51
|
Laoharawee K, Johnson MJ, Lahr WS, Sipe CJ, Kleinboehl E, Peterson JJ, Lonetree CL, Bell JB, Slipek NJ, Crane AT, Webber BR, Moriarity BS. A Pan-RNase Inhibitor Enabling CRISPR-mRNA Platforms for Engineering of Primary Human Monocytes. Int J Mol Sci 2022; 23:9749. [PMID: 36077152 PMCID: PMC9456164 DOI: 10.3390/ijms23179749] [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: 08/01/2022] [Revised: 08/16/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
Monocytes and their downstream effectors are critical components of the innate immune system. Monocytes are equipped with chemokine receptors, allowing them to migrate to various tissues, where they can differentiate into macrophage and dendritic cell subsets and participate in tissue homeostasis, infection, autoimmune disease, and cancer. Enabling genome engineering in monocytes and their effector cells will facilitate a myriad of applications for basic and translational research. Here, we demonstrate that CRISPR-Cas9 RNPs can be used for efficient gene knockout in primary human monocytes. In addition, we demonstrate that intracellular RNases are likely responsible for poor and heterogenous mRNA expression as incorporation of pan-RNase inhibitor allows efficient genome engineering following mRNA-based delivery of Cas9 and base editor enzymes. Moreover, we demonstrate that CRISPR-Cas9 combined with an rAAV vector DNA donor template mediates site-specific insertion and expression of a transgene in primary human monocytes. Finally, we demonstrate that SIRPa knock-out monocyte-derived macrophages have enhanced activity against cancer cells, highlighting the potential for application in cellular immunotherapies.
Collapse
Affiliation(s)
- Kanut Laoharawee
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Matthew J. Johnson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Walker S. Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher J. Sipe
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Evan Kleinboehl
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph J. Peterson
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Cara-lin Lonetree
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jason B. Bell
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Nicholas J. Slipek
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Andrew T. Crane
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Beau R. Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Branden S. Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
52
|
Inoue Y, Liao CW, Tsunakawa Y, Tsai IL, Takahashi S, Hamada M. Macrophage-Specific, Mafb-Deficient Mice Showed Delayed Skin Wound Healing. Int J Mol Sci 2022; 23:9346. [PMID: 36012611 PMCID: PMC9409077 DOI: 10.3390/ijms23169346] [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: 07/29/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/20/2022] Open
Abstract
Macrophages play essential roles throughout the wound repair process. Nevertheless, mechanisms regulating the process are poorly understood. MAFB is specifically expressed in the macrophages in hematopoietic tissue and is vital to homeostatic function. Comparison of the skin wound repair rates in macrophage-specific, MAFB-deficient mice (Mafbf/f::LysM-Cre) and control mice (Mafbf/f) showed that wound healing was significantly delayed in the former. For wounded GFP knock-in mice with GFP inserts in the Mafb locus, flow cytometry revealed that their GFP-positive cells expressed macrophage markers. Thus, macrophages express Mafb at wound sites. Immunohistochemical (IHC) staining, proteome analysis, and RT-qPCR of the wound tissue showed relative downregulation of Arg1, Ccl12, and Ccl2 in Mafbf/f::LysM-Cre mice. The aforementioned genes were also downregulated in the bone marrow-derived, M2-type macrophages of Mafbf/f::LysM-Cre mice. Published single-cell RNA-Seq analyses showed that Arg1, Ccl2, Ccl12, and Il-10 were expressed in distinct populations of MAFB-expressing cells. Hence, the MAFB-expressing macrophage population is heterogeneous. MAFB plays the vital role of regulating multiple genes implicated in wound healing, which suggests that MAFB is a potential therapeutic target in wound healing.
Collapse
Affiliation(s)
- Yuri Inoue
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Ching-Wei Liao
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Yuki Tsunakawa
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - I-Lin Tsai
- Global Innovation Joint-Degree Program, International Joint Degree Master’s Program, Agro-Biomedical Science in Food and Health, College of Medicine, National Taiwan University (NTU GIP-TRIAD), No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| | - Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Ibaraki, Japan
| |
Collapse
|
53
|
Zhou W, Gao F, Romero-Wolf M, Jo S, Rothenberg EV. Single-cell deletion analyses show control of pro-T cell developmental speed and pathways by Tcf7, Spi1, Gata3, Bcl11a, Erg, and Bcl11b. Sci Immunol 2022; 7:eabm1920. [PMID: 35594339 PMCID: PMC9273332 DOI: 10.1126/sciimmunol.abm1920] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
As early T cell precursors transition from multipotentiality to T lineage commitment, they change expression of multiple transcription factors. It is unclear whether individual transcription factors directly control choices between T cell identity and some alternative fate or whether these factors mostly affect proliferation or survival during the normal commitment process. Here, we unraveled the impacts of deleting individual transcription factors at two stages in early T cell development, using synchronized in vitro differentiation systems, single-cell RNA-seq with batch indexing, and controlled gene-disruption strategies. First, using a customized method for single-cell CRISPR disruption, we defined how the early-acting transcription factors Bcl11a, Erg, Spi1 (PU.1), Gata3, and Tcf7 (TCF1) function before commitment. The results revealed a kinetic tug of war within individual cells between T cell factors Tcf7 and Gata3 and progenitor factors Spi1 and Bcl11a, with an unexpected guidance role for Erg. Second, we tested how activation of transcription factor Bcl11b during commitment altered ongoing cellular programs. In knockout cells where Bcl11b expression was prevented, the cells did not undergo developmental arrest, instead following an alternative path as T lineage commitment was blocked. A stepwise, time-dependent regulatory cascade began with immediate-early transcription factor activation and E protein inhibition, finally leading Bcl11b knockout cells toward exit from the T cell pathway. Last, gene regulatory networks of transcription factor cross-regulation were extracted from the single-cell transcriptome results, characterizing the specification network operating before T lineage commitment and revealing its links to both the Bcl11b knockout alternative network and the network consolidating T cell identity during commitment.
Collapse
Affiliation(s)
- Wen Zhou
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Program in Biochemistry and Molecular Biophysics, California Institute of Technology
- Current address: BillionToOne, Menlo Park, CA
| | - Fan Gao
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Caltech Bioinformatics Resource Center, Beckman Institute of Caltech
| | - Maile Romero-Wolf
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Current address: Center for Stem Cell Biology and Regenerative Medicine, University of Southern California
| | - Suin Jo
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Current address: Washington University of St. Louis
| | - Ellen V. Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
| |
Collapse
|
54
|
Chen C, Man N, Liu F, Martin GM, Itonaga H, Sun J, Nimer SD. Epigenetic and transcriptional regulation of innate immunity in cancer. Cancer Res 2022; 82:2047-2056. [PMID: 35320354 DOI: 10.1158/0008-5472.can-21-3503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/16/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022]
Abstract
Innate immune cells participate in the detection of tumor cells via complex signaling pathways mediated by pattern-recognition receptors, such as Toll-like receptors (TLR) and NOD-like receptors (NLR). These pathways are finely tuned via multiple mechanisms, including epigenetic regulation. It is well established that hematopoietic progenitors generate innate immune cells that can regulate cancer cell behavior, and the disruption of normal hematopoiesis in pathologic states may lead to altered immunity and the development of cancer. In this review, we discuss the epigenetic and transcriptional mechanisms that underlie the initiation and amplification of innate immune signaling in cancer. We also discuss new targeting possibilities for cancer control that exploit innate immune cells and signaling molecules, potentially heralding the next generation of immunotherapy.
Collapse
Affiliation(s)
- Chuan Chen
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Na Man
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Fan Liu
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, Florida
| | - Gloria Mas Martin
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Hidehiro Itonaga
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Jun Sun
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
55
|
Subramanian S, Busch CJL, Molawi K, Geirsdottir L, Maurizio J, Vargas Aguilar S, Belahbib H, Gimenez G, Yuda RAA, Burkon M, Favret J, Gholamhosseinian Najjar S, de Laval B, Kandalla PK, Sarrazin S, Alexopoulou L, Sieweke MH. Long-term culture-expanded alveolar macrophages restore their full epigenetic identity after transfer in vivo. Nat Immunol 2022; 23:458-468. [PMID: 35210623 DOI: 10.1038/s41590-022-01146-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 01/20/2022] [Indexed: 12/31/2022]
Abstract
Alveolar macrophages (AMs) are lung tissue-resident macrophages that can be expanded in culture, but it is unknown to what extent culture affects their in vivo identity. Here we show that mouse long-term ex vivo expanded AMs (exAMs) maintained a core AM gene expression program, but showed culture adaptations related to adhesion, metabolism and proliferation. Upon transplantation into the lung, exAMs reacquired full transcriptional and epigenetic AM identity, even after several months in culture and could self-maintain long-term in the alveolar niche. Changes in open chromatin regions observed in culture were fully reversible in transplanted exAMs and resulted in a gene expression profile indistinguishable from resident AMs. Our results indicate that long-term proliferation of AMs in culture did not compromise cellular identity in vivo. The robustness of exAM identity provides new opportunities for mechanistic analysis and highlights the therapeutic potential of exAMs.
Collapse
Affiliation(s)
- Sethuraman Subramanian
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.,Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France.,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), Berlin, Germany
| | - Clara Jana-Lui Busch
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Kaaweh Molawi
- Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France.,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), Berlin, Germany
| | | | | | - Stephanie Vargas Aguilar
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.,Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France.,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), Berlin, Germany
| | | | - Gregory Gimenez
- Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France
| | - Ridzky Anis Advent Yuda
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Michaela Burkon
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Jérémy Favret
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | | | | | - Prashanth Kumar Kandalla
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Sandrine Sarrazin
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.,Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France
| | | | - Michael H Sieweke
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany. .,Aix-Marseille University, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
56
|
Domanska D, Majid U, Karlsen VT, Merok MA, Beitnes ACR, Yaqub S, Bækkevold ES, Jahnsen FL. Single-cell transcriptomic analysis of human colonic macrophages reveals niche-specific subsets. J Exp Med 2022; 219:212998. [PMID: 35139155 PMCID: PMC8932544 DOI: 10.1084/jem.20211846] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/17/2022] Open
Abstract
Macrophages are a heterogeneous population of cells involved in tissue homeostasis, inflammation, and cancer. Although macrophages are densely distributed throughout the human intestine, our understanding of how gut macrophages maintain tissue homeostasis is limited. Here we show that colonic lamina propria macrophages (LpMs) and muscularis macrophages (MMs) consist of monocyte-like cells that differentiate into multiple transcriptionally distinct subsets. LpMs comprise subsets with proinflammatory properties and subsets with high antigen-presenting and phagocytic capacity. The latter are strategically positioned close to the surface epithelium. Most MMs differentiate along two trajectories: one that upregulates genes associated with immune activation and angiogenesis, and one that upregulates genes associated with neuronal homeostasis. Importantly, MMs are located adjacent to neurons and vessels. Cell–cell interaction and gene network analysis indicated that survival, migration, transcriptional reprogramming, and niche-specific localization of LpMs and MMs are controlled by an extensive interaction with tissue-resident cells and a few key transcription factors.
Collapse
Affiliation(s)
- Diana Domanska
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Umair Majid
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Victoria T Karlsen
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marianne A Merok
- Department of Gastrointestinal Surgery, Akershus University Hospital, Lørenskog, Norway
| | | | - Sheraz Yaqub
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Hepatobiliary Surgery, Oslo University Hospital, Oslo, Norway
| | - Espen S Bækkevold
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Institute of Oral Biology, University of Oslo, Oslo, Norway
| | - Frode L Jahnsen
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
57
|
Evren E, Ringqvist E, Doisne JM, Thaller A, Sleiers N, Flavell RA, Di Santo JP, Willinger T. CD116+ fetal precursors migrate to the perinatal lung and give rise to human alveolar macrophages. J Exp Med 2022; 219:212959. [PMID: 35019940 PMCID: PMC8759608 DOI: 10.1084/jem.20210987] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 11/05/2021] [Accepted: 12/13/2021] [Indexed: 12/27/2022] Open
Abstract
Despite their importance in lung health and disease, it remains unknown how human alveolar macrophages develop early in life. Here we define the ontogeny of human alveolar macrophages from embryonic progenitors in vivo, using a humanized mouse model expressing human cytokines (MISTRG mice). We identified alveolar macrophage progenitors in human fetal liver that expressed the GM-CSF receptor CD116 and the transcription factor MYB. Transplantation experiments in MISTRG mice established a precursor-product relationship between CD34-CD116+ fetal liver cells and human alveolar macrophages in vivo. Moreover, we discovered circulating CD116+CD64-CD115+ macrophage precursors that migrated from the liver to the lung. Similar precursors were present in human fetal lung and expressed the chemokine receptor CX3CR1. Fetal CD116+CD64- macrophage precursors had a proliferative gene signature, outcompeted adult precursors in occupying the perinatal alveolar niche, and developed into functional alveolar macrophages. The discovery of the fetal alveolar macrophage progenitor advances our understanding of human macrophage origin and ontogeny.
Collapse
Affiliation(s)
- Elza Evren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Emma Ringqvist
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jean-Marc Doisne
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut national de la santé et de la recherche médicale U1223, Paris, France
| | - Anna Thaller
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut national de la santé et de la recherche médicale U1223, Paris, France.,Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Natalie Sleiers
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT.,Howard Hughes Medical Institute, Chevy Chase, MD
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France.,Institut national de la santé et de la recherche médicale U1223, Paris, France
| | - Tim Willinger
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
58
|
Dastagir N, Beal Z, Godwin J. Tissue origin of cytotoxic natural killer cells dictates their differential roles in mouse digit tip regeneration and progenitor cell survival. Stem Cell Reports 2022; 17:633-648. [PMID: 35120621 PMCID: PMC9039750 DOI: 10.1016/j.stemcr.2022.01.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/06/2022] [Accepted: 01/07/2022] [Indexed: 02/08/2023] Open
Abstract
Regeneration of amputated digit tips relies on mesenchymal progenitor cells and their differentiation into replacement bone and tissue stroma. Natural killer (NK) cells have well-characterized roles in antigen-independent killing of virally infected, pre-tumorous, or stressed cells; however, the potential for cytotoxic activity against regenerative progenitor cells is unclear. We identified NK cell recruitment to the regenerating digit tip, and NK cytotoxicity was observed against osteoclast and osteoblast progenitors. Adoptive cell transplants of spleen NK (SpNK) or thymus NK (ThNK) donor cells into immunodeficient mice demonstrated ThNK cell-induced apoptosis with a reduction in osteoclasts, osteoblasts, and proliferative cells, resulting in inhibition of regeneration. Adoptive transfer of NK cells deficient in NK cell activation genes identified that promotion of regeneration by SpNK cells requires Ncr1, whereas inhibition by ThNK cells is mediated via Klrk1 and perforin. Successful future therapies aimed at enhancing regeneration will require a deeper understanding of progenitor cell protection from NK cell cytotoxicity.
Collapse
Affiliation(s)
- Nadjib Dastagir
- The Jackson Laboratory, Bar Harbor, ME 04609, USA,Mount Desert Island Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Salisbury Cove, ME 04609, USA,Medical School of Hanover, 30659 Hannover, Germany
| | - Zachery Beal
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - James Godwin
- The Jackson Laboratory, Bar Harbor, ME 04609, USA,Mount Desert Island Biological Laboratory, Kathryn W. Davis Center for Regenerative Biology and Aging, Salisbury Cove, ME 04609, USA,Corresponding author
| |
Collapse
|
59
|
Pan Y, Cao S, Terker AS, Tang J, Sasaki K, Wang Y, Niu A, Luo W, Fan X, Wang S, Wilson MH, Zhang MZ, Harris RC. Myeloid cyclooxygenase-2/prostaglandin E2/E-type prostanoid receptor 4 promotes transcription factor MafB-dependent inflammatory resolution in acute kidney injury. Kidney Int 2022; 101:79-91. [PMID: 34774558 PMCID: PMC8741730 DOI: 10.1016/j.kint.2021.09.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/09/2021] [Accepted: 09/24/2021] [Indexed: 01/07/2023]
Abstract
Following acute injury to the kidney, macrophages play an important role in recovery of functional and structural integrity, but organ fibrosis and progressive functional decline occur with incomplete recovery. Pro-resolving macrophages are characterized by increased cyclooxygenase 2 (COX-2) expression and this expression was selectively increased in kidney macrophages following injury and myeloid-specific COX-2 deletion inhibited recovery. Deletion of the myeloid prostaglandin E2 (PGE2) receptor, E-type prostanoid receptor 4 (EP4), mimicked effects seen with myeloid COX-2-/- deletion. PGE2-mediated EP4 activation induced expression of the transcription factor MafB in kidney macrophages, which upregulated anti-inflammatory genes and suppressed pro-inflammatory genes. Myeloid Mafb deletion recapitulated the effects seen with either myeloid COX-2 or EP4 deletion following acute kidney injury, with delayed recovery, persistent presence of pro-inflammatory kidney macrophages, and increased kidney fibrosis. Thus, our studies identified a previously unknown mechanism by which prostaglandins modulate macrophage phenotype following acute organ injury and provide new insight into mechanisms underlying detrimental kidney effects of non-steroidal anti-inflammatory drugs that inhibit cyclooxygenase activity.
Collapse
Affiliation(s)
- Yu Pan
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Division of Nephrology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shirong Cao
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Andrew S Terker
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Jiaqi Tang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Kensuke Sasaki
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Yinqiu Wang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Aolei Niu
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Wentian Luo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Xiaofeng Fan
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Suwan Wang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Matthew H Wilson
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Vanderbilt Center for Kidney Disease, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Department of Veterans Affairs, Nashville, Tennessee, USA.
| |
Collapse
|
60
|
Zhao D, Yang F, Wang Y, Li S, Li Y, Hou F, Yang W, Liu D, Tao Y, Li Q, Wang J, He F, Tang L. ALK1 signaling is required for the homeostasis of Kupffer cells and prevention of bacterial infection. J Clin Invest 2021; 132:150489. [PMID: 34874921 PMCID: PMC8803331 DOI: 10.1172/jci150489] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 11/30/2021] [Indexed: 11/18/2022] Open
Abstract
Macrophages are highly heterogeneous immune cells that fulfill tissue-specific functions. Tissue-derived signals play a critical role in determining macrophage heterogeneity. However, these signals remain largely unknown. The BMP receptor activin receptor–like kinase 1 (ALK1) is well known for its role in blood vessel formation; however, its role within the immune system has never been revealed to our knowledge. Here, we found that BMP9/BMP10/ALK1 signaling controlled the identity and self-renewal of Kupffer cells (KCs) through a Smad4-dependent pathway. In contrast, ALK1 was dispensable for the maintenance of macrophages located in the lung, kidney, spleen, and brain. Following ALK1 deletion, KCs were lost over time and were replaced by monocyte-derived macrophages. These hepatic macrophages showed significantly reduced expression of the complement receptor VSIG4 and alterations in immune zonation and morphology, which is important for the tissue-specialized function of KCs. Furthermore, we found that this signaling pathway was important for KC-mediated Listeria monocytogenes capture, as the loss of ALK1 and Smad4 led to a failure of bacterial capture and overwhelming disseminated infections. Thus, ALK1 signaling instructs a tissue-specific phenotype that allows KCs to protect the host from systemic bacterial dissemination.
Collapse
Affiliation(s)
- Dianyuan Zhao
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Fengjiao Yang
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Yang Wang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Site Li
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Yang Li
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Fei Hou
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Wenting Yang
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Di Liu
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Yuandong Tao
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Qian Li
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Jing Wang
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fuchu He
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| | - Li Tang
- Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing, China
| |
Collapse
|
61
|
Belhocine S, Machado Xavier A, Distéfano-Gagné F, Fiola S, Rivest S, Gosselin D. Context-dependent transcriptional regulation of microglial proliferation. Glia 2021; 70:572-589. [PMID: 34862814 DOI: 10.1002/glia.24124] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 01/12/2023]
Abstract
Microglia proliferate during brain development and brain lesions, but how this is coordinated at the transcriptional level is not well understood. Here, we investigated fundamental aspects of the transcriptional process associated with proliferation of mouse microglia during postnatal development and in adults in a model of induced microglial depletion-repopulation. While each proliferative subset displayed globally a distinct signature of gene expression, they also co-expressed a subgroup of 1370 genes at higher levels than quiescent microglia. Expression of these may be coordinated by one of two mechanisms of regulation with distinct properties. A first mechanism augments expression of genes already expressed in quiescent microglia and is subject to regulation by Klf/Sp, Nfy, and Ets transcription factors. Alternatively, a second mechanism enables de novo transcription of cell cycle genes and requires additional regulatory input from Lin54 and E2f transcription factors. Of note, transcriptional upregulation of E2f1 and E2f2 family members may represent a critical regulatory checkpoint to enable microglia to achieve efficient cell cycling. Furthermore, analysis of the activity profile of the repertoire of promoter-distal genomic regulatory elements suggests a relatively restricted role for these elements in coordinating cell cycle gene expression in microglia. Overall, proliferating microglia integrates regulation of cell cycle gene expression with their broader, context-dependent, transcriptional landscape.
Collapse
Affiliation(s)
- Sarah Belhocine
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada.,Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Canada
| | - André Machado Xavier
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada.,Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Canada
| | - Félix Distéfano-Gagné
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada.,Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Canada
| | - Stéphanie Fiola
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada
| | - Serge Rivest
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada.,Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Canada
| | - David Gosselin
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Québec, Canada.,Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Canada
| |
Collapse
|
62
|
Wessendarp M, Watanabe-Chailland M, Liu S, Stankiewicz T, Ma Y, Kasam RK, Shima K, Chalk C, Carey B, Rosendale LR, Dominique Filippi M, Arumugam P. Role of GM-CSF in regulating metabolism and mitochondrial functions critical to macrophage proliferation. Mitochondrion 2021; 62:85-101. [PMID: 34740864 DOI: 10.1016/j.mito.2021.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 10/14/2021] [Accepted: 10/28/2021] [Indexed: 12/14/2022]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF) exerts pleiotropic effects on macrophages and is required for self-renewal but the mechanisms responsible are unknown. Using mouse models with disrupted GM-CSF signaling, we show GM-CSF is critical for mitochondrial turnover, functions, and integrity. GM-CSF signaling is essential for fatty acid β-oxidation and markedly increased tricarboxylic acid cycle activity, oxidative phosphorylation, and ATP production. GM-CSF also regulated cytosolic pathways including glycolysis, pentose phosphate pathway, and amino acid synthesis. We conclude that GM-CSF regulates macrophages in part through a critical role in maintaining mitochondria, which are necessary for cellular metabolism as well as proliferation and self-renewal.
Collapse
Affiliation(s)
- Matthew Wessendarp
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | | | - Serena Liu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Yan Ma
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | | | - Kenjiro Shima
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | - Claudia Chalk
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | - Brenna Carey
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA
| | | | | | - Paritha Arumugam
- Translational Pulmonary Science Center, Children's Hospital Medical Center (CCHMC), Cincinnati, OH, USA; Division of Pulmonary Biology, CCHMC, OH, USA.
| |
Collapse
|
63
|
Moura Silva H, Kitoko JZ, Queiroz CP, Kroehling L, Matheis F, Yang KL, Reis BS, Ren-Fielding C, Littman DR, Bozza MT, Mucida D, Lafaille JJ. c-MAF-dependent perivascular macrophages regulate diet-induced metabolic syndrome. Sci Immunol 2021; 6:eabg7506. [PMID: 34597123 DOI: 10.1126/sciimmunol.abg7506] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hernandez Moura Silva
- Kimmel Center for Biology and Medicine at the Skirball Institute; New York University School of Medicine, New York, NY 10016, USA
| | - Jamil Zola Kitoko
- Kimmel Center for Biology and Medicine at the Skirball Institute; New York University School of Medicine, New York, NY 10016, USA.,Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Camila Pereira Queiroz
- Kimmel Center for Biology and Medicine at the Skirball Institute; New York University School of Medicine, New York, NY 10016, USA.,Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas. Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Lina Kroehling
- Kimmel Center for Biology and Medicine at the Skirball Institute; New York University School of Medicine, New York, NY 10016, USA
| | - Fanny Matheis
- Laboratory of Mucosal Immunology, Rockefeller University, New York, NY 10065, USA
| | - Katharine Lu Yang
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Bernardo S Reis
- Laboratory of Mucosal Immunology, Rockefeller University, New York, NY 10065, USA
| | | | - Dan R Littman
- Kimmel Center for Biology and Medicine at the Skirball Institute; New York University School of Medicine, New York, NY 10016, USA.,Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA.,Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Marcelo Torres Bozza
- Laboratório de Inflamação e Imunidade, Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, Rockefeller University, New York, NY 10065, USA
| | - Juan J Lafaille
- Kimmel Center for Biology and Medicine at the Skirball Institute; New York University School of Medicine, New York, NY 10016, USA.,Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| |
Collapse
|
64
|
Affiliation(s)
- Gwendalyn J Randolph
- Department of Pathology & Immunology, Washington University, St. Louis, MO, USA.
| |
Collapse
|
65
|
Zhan Y, Zhang Y, Zhang S, Coughlan H, Baldoni PL, Jacquelot N, Cao WHJ, Preston S, Louis C, Rautela J, Pellegrini M, Wicks IP, Alexander WS, Harrison LC, Lew AM, Smyth GK, Nutt SL, Chopin M. Differential requirement for the Polycomb repressor complex 2 in dendritic cell and tissue-resident myeloid cell homeostasis. Sci Immunol 2021; 6:eabf7268. [PMID: 34533976 DOI: 10.1126/sciimmunol.abf7268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Yifan Zhan
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.,Drug Discovery, Shanghai Huaota Biopharma, Shanghai, China
| | - Yuxia Zhang
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Shengbo Zhang
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Hannah Coughlan
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Pedro L Baldoni
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicolas Jacquelot
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Wang H J Cao
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Simon Preston
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Cynthia Louis
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jai Rautela
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Marc Pellegrini
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ian P Wicks
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Warren S Alexander
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Leonard C Harrison
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew M Lew
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.,Department of Microbiology and Immunology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Gordon K Smyth
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michaël Chopin
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
66
|
Bene K, Halasz L, Nagy L. Transcriptional repression shapes the identity and function of tissue macrophages. FEBS Open Bio 2021; 11:3218-3229. [PMID: 34358410 PMCID: PMC8634859 DOI: 10.1002/2211-5463.13269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/16/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
The changing extra‐ and intracellular microenvironment calls for rapid cell fate decisions that are precisely and primarily regulated at the transcriptional level. The cellular components of the immune system are excellent examples of how cells respond and adapt to different environmental stimuli. Innate immune cells such as macrophages are able to modulate their transcriptional programs and epigenetic regulatory networks through activation and repression of particular genes, allowing them to quickly respond to a rapidly changing environment. Tissue macrophages are essential components of different immune‐ and nonimmune cell‐mediated physiological mechanisms in mammals and are widely used models for investigating transcriptional regulatory mechanisms. Therefore, it is critical to unravel the distinct sets of transcription activators, repressors, and coregulators that play roles in determining tissue macrophage identity and functions during homeostasis, as well as in diseases affecting large human populations, such as metabolic syndromes, immune‐deficiencies, and tumor development. In this review, we will focus on transcriptional repressors that play roles in tissue macrophage development and function under physiological conditions.
Collapse
Affiliation(s)
- Krisztian Bene
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Laszlo Halasz
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| |
Collapse
|
67
|
Baasch S, Giansanti P, Kolter J, Riedl A, Forde AJ, Runge S, Zenke S, Elling R, Halenius A, Brabletz S, Hengel H, Kuster B, Brabletz T, Cicin-Sain L, Arens R, Vlachos A, Rohr JC, Stemmler MP, Kopf M, Ruzsics Z, Henneke P. Cytomegalovirus subverts macrophage identity. Cell 2021; 184:3774-3793.e25. [PMID: 34115982 DOI: 10.1016/j.cell.2021.05.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/26/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022]
Abstract
Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.
Collapse
Affiliation(s)
- Sebastian Baasch
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Piero Giansanti
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Julia Kolter
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - André Riedl
- Institute of Virology, University Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Aaron James Forde
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Solveig Runge
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Simon Zenke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Roland Elling
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Center for Pediatrics and Adolescent Medicine, University Medical Center, 79106 Freiburg, Germany
| | - Anne Halenius
- Institute of Virology, University Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Simone Brabletz
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Hartmut Hengel
- Institute of Virology, University Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany; Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), Technical University Munich, 85354 Freising, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Luka Cicin-Sain
- Immune Aging and Chronic Infections Group, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hanover Medical School (MHH), 30625 Hanover, Germany
| | - Ramon Arens
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Andreas Vlachos
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany; Center for Basics in Neuromodulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Jan Christopher Rohr
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Center for Pediatrics and Adolescent Medicine, University Medical Center, 79106 Freiburg, Germany
| | - Marc Philippe Stemmler
- Department of Experimental Medicine I, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich Alexander University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Manfred Kopf
- Institute of Molecular Health Sciences, ETH Zürich, 8093 Zürich, Switzerland
| | - Zsolt Ruzsics
- Institute of Virology, University Medical Center, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
| | - Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Center for Pediatrics and Adolescent Medicine, University Medical Center, 79106 Freiburg, Germany.
| |
Collapse
|
68
|
Li SY, Gu X, Heinrich A, Hurley EG, Capel B, DeFalco T. Loss of Mafb and Maf distorts myeloid cell ratios and disrupts fetal mouse testis vascularization and organogenesis†. Biol Reprod 2021; 105:958-975. [PMID: 34007995 DOI: 10.1093/biolre/ioab098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022] Open
Abstract
Testis differentiation is initiated when Sry in pre-Sertoli cells directs the gonad toward a male-specific fate. Sertoli cells are essential for testis development, but cell types within the interstitial compartment, such as immune and endothelial cells, are also critical for organ formation. Our previous work implicated macrophages in fetal testis morphogenesis, but little is known about genes underlying immune cell development during organogenesis. Here we examine the role of the immune-associated genes Mafb and Maf in mouse fetal gonad development, and we demonstrate that deletion of these genes leads to aberrant hematopoiesis manifested by supernumerary gonadal monocytes. Mafb; Maf double knockout embryos underwent initial gonadal sex determination normally, but exhibited testicular hypervascularization, testis cord formation defects, Leydig cell deficit, and a reduced number of germ cells. In general, Mafb and Maf alone were dispensable for gonad development; however, when both genes were deleted, we observed significant defects in testicular morphogenesis, indicating that Mafb and Maf work redundantly during testis differentiation. These results demonstrate previously unappreciated roles for Mafb and Maf in immune and vascular development and highlight the importance of interstitial cells in gonadal differentiation.
Collapse
Affiliation(s)
- Shu-Yun Li
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xiaowei Gu
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Anna Heinrich
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Emily G Hurley
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA.,Department of Obstetrics and Gynecology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Blanche Capel
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Tony DeFalco
- Division of Reproductive Sciences, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| |
Collapse
|
69
|
Takahashi S. Functional analysis of large MAF transcription factors and elucidation of their relationships with human diseases. Exp Anim 2021; 70:264-271. [PMID: 33762508 PMCID: PMC8390310 DOI: 10.1538/expanim.21-0027] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The large MAF transcription factor group is a group of transcription factors with an acidic region, a basic region, and a leucine zipper region. Four types of MAF, MAFA, MAFB, c-MAF, and NRL, have been identified in humans and mice. In order to elucidate the functions of the large MAF transcription factor group in vivo, our research group created genetically modified MAFA-, MAFB-, and c-MAF-deficient mice and analyzed their phenotypes. MAFA is expressed in pancreatic β cells and is essential for insulin transcription and secretion. MAFB is essential for the development of pancreatic endocrine cells, formation of inner ears, podocyte function in the kidneys, and functional differentiation of macrophages. c-MAF is essential for lens formation and osteoblast differentiation. Furthermore, a single-base mutation in genes encoding the large MAF transcription factor group causes congenital renal disease, eye disease, bone disease, diabetes, and tumors in humans. This review describes the functions of large MAF transcription factors in vivo and their relationships with human diseases.
Collapse
Affiliation(s)
- Satoru Takahashi
- Department of Anatomy and Embryology, Laboratory Animal Resource Center in Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
70
|
Han Y, Shao W, Zhong D, Ma C, Wei X, Ahmed A, Yu T, Jing W, Jing L. Zebrafish mafbb Mutants Display Osteoclast Over-Activation and Bone Deformity Resembling Osteolysis in MCTO Patients. Biomolecules 2021; 11:biom11030480. [PMID: 33806930 PMCID: PMC8004647 DOI: 10.3390/biom11030480] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/31/2022] Open
Abstract
Multicentric carpotarsal osteolysis (MCTO) is a rare skeletal dysplasia with osteolysis at the carpal and tarsal bones. Heterozygous missense mutations in the transcription factor MAFB are found in patients with MCTO. MAFB is reported to negatively regulate osteoclastogenesis in vitro. However, the in vivo function of MAFB and its relation to MCTO remains unknown. In this study, we generated zebrafish MAFB homolog mafbb mutant utilizing CRISPR/Cas9 technology. Mafbb deficient zebrafish demonstrated enhanced osteoclast cell differentiation and abnormal cartilage and bone development resembling MCTO patients. It is known that osteoclasts are hematopoietic cells derived from macrophages. Loss of mafbb caused selective expansion of definitive macrophages and myeloid cells, supporting that mafbb restricts myeloid differentiation in vivo. We also demonstrate that MAFB MCTO mutations failed to rescue the defective osteoclastogenesis in mafbb-/- embryos, but did not affect osteoclast cells in wild type embryos. The mechanism of MCTO mutations is likely haploinsufficiency. Zebrafish mafbb mutant provides a useful model to study the function of MAFB in osteoclastogenesis and the related MCTO disease.
Collapse
Affiliation(s)
- Yujie Han
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.H.); (W.S.); (D.Z.); (C.M.); (X.W.); (A.A.)
| | - Weihao Shao
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.H.); (W.S.); (D.Z.); (C.M.); (X.W.); (A.A.)
| | - Dan Zhong
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.H.); (W.S.); (D.Z.); (C.M.); (X.W.); (A.A.)
| | - Cui Ma
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.H.); (W.S.); (D.Z.); (C.M.); (X.W.); (A.A.)
| | - Xiaona Wei
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.H.); (W.S.); (D.Z.); (C.M.); (X.W.); (A.A.)
| | - Abrar Ahmed
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.H.); (W.S.); (D.Z.); (C.M.); (X.W.); (A.A.)
| | - Tingting Yu
- Shanghai Children’s Medical Center, Department of Medical Genetics and Molecular Diagnostic Laboratory, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China;
| | - Wei Jing
- Department of Hepatobiliary Pancreatic Surgery, Shanghai Changhai Hospital, Shanghai 200433, China
- Correspondence: (W.J.); (L.J.)
| | - Lili Jing
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; (Y.H.); (W.S.); (D.Z.); (C.M.); (X.W.); (A.A.)
- Correspondence: (W.J.); (L.J.)
| |
Collapse
|
71
|
Ratnayake D, Nguyen PD, Rossello FJ, Wimmer VC, Tan JL, Galvis LA, Julier Z, Wood AJ, Boudier T, Isiaku AI, Berger S, Oorschot V, Sonntag C, Rogers KL, Marcelle C, Lieschke GJ, Martino MM, Bakkers J, Currie PD. Macrophages provide a transient muscle stem cell niche via NAMPT secretion. Nature 2021; 591:281-287. [PMID: 33568815 DOI: 10.1038/s41586-021-03199-7] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/07/2021] [Indexed: 01/30/2023]
Abstract
Skeletal muscle regenerates through the activation of resident stem cells. Termed satellite cells, these normally quiescent cells are induced to proliferate by wound-derived signals1. Identifying the source and nature of these cues has been hampered by an inability to visualize the complex cell interactions that occur within the wound. Here we use muscle injury models in zebrafish to systematically capture the interactions between satellite cells and the innate immune system after injury, in real time, throughout the repair process. This analysis revealed that a specific subset of macrophages 'dwell' within the injury, establishing a transient but obligate niche for stem cell proliferation. Single-cell profiling identified proliferative signals that are secreted by dwelling macrophages, which include the cytokine nicotinamide phosphoribosyltransferase (Nampt, which is also known as visfatin or PBEF in humans). Nampt secretion from the macrophage niche is required for muscle regeneration, acting through the C-C motif chemokine receptor type 5 (Ccr5), which is expressed on muscle stem cells. This analysis shows that in addition to their ability to modulate the immune response, specific macrophage populations also provide a transient stem-cell-activating niche, directly supplying proliferation-inducing cues that govern the repair process that is mediated by muscle stem cells. This study demonstrates that macrophage-derived niche signals for muscle stem cells, such as NAMPT, can be applied as new therapeutic modalities for skeletal muscle injury and disease.
Collapse
Affiliation(s)
- Dhanushika Ratnayake
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Phong D Nguyen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Fernando J Rossello
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,University of Melbourne Centre for Cancer Research, The University of Melbourne, Melbourne, Victoria, Australia
| | - Verena C Wimmer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jean L Tan
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Laura A Galvis
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,Institut NeuroMyoGène (INMG), University Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, France
| | - Ziad Julier
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Alasdair J Wood
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Thomas Boudier
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Abdulsalam I Isiaku
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Silke Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Viola Oorschot
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Melbourne, Victoria, Australia.,European Molecular Biology Laboratory, Electron Microscopy Core Facility, Heidelberg, Germany
| | - Carmen Sonntag
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Christophe Marcelle
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,Institut NeuroMyoGène (INMG), University Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Lyon, France
| | - Graham J Lieschke
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Mikaël M Martino
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia.,EMBL Australia, Monash University, Clayton, Victoria, Australia
| | - Jeroen Bakkers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia. .,EMBL Australia, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
72
|
Wang J, Wang Y, Chu Y, Li Z, Yu X, Huang Z, Xu J, Zheng L. Tumor-derived adenosine promotes macrophage proliferation in human hepatocellular carcinoma. J Hepatol 2021; 74:627-637. [PMID: 33137360 DOI: 10.1016/j.jhep.2020.10.021] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Macrophages (Mϕ) represent a major component of tumor tissues and play an important role in both tumor progression and therapeutic response. Although tumor Mϕ are generally considered to be derived from circulating monocytes, emerging evidence indicates that tissue Mϕ pools can be maintained by self-renewal. We aimed to elucidate the contribution, phenotype, and regulatory mechanisms of proliferating Mϕ in human hepatocellular carcinoma (HCC). METHODS Flow cytometry analyses were performed to examine the presence and phenotype of proliferating Mϕ in fresh HCC tissues. Dual immunofluorescence staining was applied to analyze the prognostic value of proliferating Mϕ. The underlying regulatory mechanisms were examined using human monocyte-derived Mϕ. RESULTS Tumor-infiltrating Mϕ exhibited a significantly higher proliferative capacity than Mϕ in non-tumor tissues. A higher level of Mϕ proliferation was positively correlated with Mϕ density in the tumor and a poor prognosis in patients with HCC. Proliferating Mϕ were less differentiated (with increased CD206 expression) and were induced by the tumor cell-derived soluble small molecule, adenosine, but not proteins, lipids, or large peptides. Mechanistic studies demonstrated that autocrine granulocyte-macrophage colony-stimulating factor (GM-CSF) released by tumor-stimulated Mϕ could enhance A2A receptor expression on Mϕ and function synergistically with adenosine to elicit Mϕ proliferation in HCC. CONCLUSIONS Local Mϕ proliferation is an important mechanism for Mϕ accumulation in HCC tissues. Tumor-derived adenosine functions synergistically with autocrine GM-CSF released from activated Mϕ, which promotes Mϕ proliferation. Thus, selective modulation of Mϕ accumulation at the source may provide a novel strategy for cancer therapy. LAY SUMMARY Tumor-associated macrophages (TAMs) have been reported to play an essential role in both tumor progression and therapeutic response. A fundamental understanding of the mechanisms that regulate macrophage accumulation in tumors will undoubtedly lead to the development of strategies to target macrophages with high specificity and efficiency. The current study unveils a novel mechanism by which local proliferation is linked to macrophage accumulation in the tumor milieu, identifying potential targets for future immune-based anticancer therapies.
Collapse
Affiliation(s)
- Junfeng Wang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Yongchun Wang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Yifan Chu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Zhixiong Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Xingjuan Yu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Zhijie Huang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Jing Xu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Limin Zheng
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China; MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China.
| |
Collapse
|
73
|
Bell S, Rigas AS, Magnusson MK, Ferkingstad E, Allara E, Bjornsdottir G, Ramond A, Sørensen E, Halldorsson GH, Paul DS, Burgdorf KS, Eggertsson HP, Howson JMM, Thørner LW, Kristmundsdottir S, Astle WJ, Erikstrup C, Sigurdsson JK, Vuckovic D, Dinh KM, Tragante V, Surendran P, Pedersen OB, Vidarsson B, Jiang T, Paarup HM, Onundarson PT, Akbari P, Nielsen KR, Lund SH, Juliusson K, Magnusson MI, Frigge ML, Oddsson A, Olafsson I, Kaptoge S, Hjalgrim H, Runarsson G, Wood AM, Jonsdottir I, Hansen TF, Sigurdardottir O, Stefansson H, Rye D, Peters JE, Westergaard D, Holm H, Soranzo N, Banasik K, Thorleifsson G, Ouwehand WH, Thorsteinsdottir U, Roberts DJ, Sulem P, Butterworth AS, Gudbjartsson DF, Danesh J, Brunak S, Di Angelantonio E, Ullum H, Stefansson K. A genome-wide meta-analysis yields 46 new loci associating with biomarkers of iron homeostasis. Commun Biol 2021; 4:156. [PMID: 33536631 PMCID: PMC7859200 DOI: 10.1038/s42003-020-01575-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023] Open
Abstract
Iron is essential for many biological functions and iron deficiency and overload have major health implications. We performed a meta-analysis of three genome-wide association studies from Iceland, the UK and Denmark of blood levels of ferritin (N = 246,139), total iron binding capacity (N = 135,430), iron (N = 163,511) and transferrin saturation (N = 131,471). We found 62 independent sequence variants associating with iron homeostasis parameters at 56 loci, including 46 novel loci. Variants at DUOX2, F5, SLC11A2 and TMPRSS6 associate with iron deficiency anemia, while variants at TF, HFE, TFR2 and TMPRSS6 associate with iron overload. A HBS1L-MYB intergenic region variant associates both with increased risk of iron overload and reduced risk of iron deficiency anemia. The DUOX2 missense variant is present in 14% of the population, associates with all iron homeostasis biomarkers, and increases the risk of iron deficiency anemia by 29%. The associations implicate proteins contributing to the main physiological processes involved in iron homeostasis: iron sensing and storage, inflammation, absorption of iron from the gut, iron recycling, erythropoiesis and bleeding/menstruation.
Collapse
Affiliation(s)
- Steven Bell
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Andreas S Rigas
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Magnus K Magnusson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| | | | - Elias Allara
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | | | - Anna Ramond
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Dirk S Paul
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Kristoffer S Burgdorf
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Joanna M M Howson
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Lise W Thørner
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - William J Astle
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Medical Research Council Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Dragana Vuckovic
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Khoa M Dinh
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Vinicius Tragante
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Rutherford Fund Fellow, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Ole B Pedersen
- Department of Clinical Immunology, Næstved Hospital, Næstved, Denmark
| | | | - Tao Jiang
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Helene M Paarup
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Pall T Onundarson
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
- Department of Laboratory Hematology, Landspitali, the National University Hospital of Iceland, Reykjavik, Iceland
| | - Parsa Akbari
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Kaspar R Nielsen
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | | | | | | | | | | | - Isleifur Olafsson
- Department of Clinical Biochemistry, Landspitali, the National University Hospital of Iceland, Reykjavik, Iceland
| | - Stephen Kaptoge
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Henrik Hjalgrim
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
| | | | - Angela M Wood
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Ingileif Jonsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - Thomas F Hansen
- Danish Headache Center, Department of Neurology, Rigshospitalet-Glostrup, Glostrup, Denmark
- Institute of Biological Psychiatry, Copenhagen University Hospital MHC Sct. Hans, Roskilde, Denmark
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | | | | | - David Rye
- Department of Neurology and Program in Sleep, Emory University School of Medicine, Atlanta, GA, USA
| | - James E Peters
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - David Westergaard
- Translational Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hilma Holm
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
| | - Nicole Soranzo
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Karina Banasik
- Translational Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Willem H Ouwehand
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
- UK National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK
| | - Unnur Thorsteinsdottir
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| | - David J Roberts
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- Radcliffe Department of Medicine and National Health Service Blood and Transplant, John Radcliffe Hospital, Oxford, UK
- UK National Health Service Blood and Transplant, John Radcliffe Hospital, Oxford, OX3 9BQ, UK
| | | | - Adam S Butterworth
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Daniel F Gudbjartsson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland
| | - John Danesh
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Department of Human Genetics, Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Søren Brunak
- Translational Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emanuele Di Angelantonio
- The National Institute for Health Research Blood and Transplant Research Unit in Donor Health and Genomics at the University of Cambridge, University of Cambridge, Cambridge, UK.
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK.
- UK National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, UK.
| | - Henrik Ullum
- Department of Clinical Immunology, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Kari Stefansson
- deCODE genetics/Amgen Inc., Reykjavik, Iceland.
- Faculty of Medicine, School of Health Sciences, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
74
|
Vega MA, Simón-Fuentes M, González de la Aleja A, Nieto C, Colmenares M, Herrero C, Domínguez-Soto Á, Corbí ÁL. MAFB and MAF Transcription Factors as Macrophage Checkpoints for COVID-19 Severity. Front Immunol 2020; 11:603507. [PMID: 33312178 PMCID: PMC7708330 DOI: 10.3389/fimmu.2020.603507] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/19/2020] [Indexed: 01/10/2023] Open
Abstract
Defective IFN production and exacerbated inflammatory and pro-fibrotic responses are hallmarks of SARS-CoV-2 infection in severe COVID-19. Based on these hallmarks, and considering the pivotal role of macrophages in COVID-19 pathogenesis, we hypothesize that the transcription factors MAFB and MAF critically contribute to COVID-19 progression by shaping the response of macrophages to SARS-CoV-2. Our proposal stems from the recent identification of pathogenic lung macrophage subsets in severe COVID-19, and takes into consideration the previously reported ability of MAFB to dampen IFN type I production, as well as the critical role of MAFB and MAF in the acquisition and maintenance of the transcriptional signature of M-CSF-conditioned human macrophages. Solid evidences are presented that link overexpression of MAFB and silencing of MAF expression with clinical and biological features of severe COVID-19. As a whole, we propose that a high MAFB/MAF expression ratio in lung macrophages could serve as an accurate diagnostic tool for COVID-19 progression. Indeed, reversing the macrophage MAFB/MAF expression ratio might impair the exacerbated inflammatory and profibrotic responses, and restore the defective IFN type I production, thus becoming a potential strategy to limit severity of COVID-19.
Collapse
Affiliation(s)
- Miguel A. Vega
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| | | | | | | | | | | | | | - Ángel L. Corbí
- Myeloid Cell Laboratory, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| |
Collapse
|
75
|
Choudhary I, Vo T, Paudel K, Patial S, Saini Y. Compartment-specific transcriptomics of ozone-exposed murine lungs reveals sex- and cell type-associated perturbations relevant to mucoinflammatory lung diseases. Am J Physiol Lung Cell Mol Physiol 2020; 320:L99-L125. [PMID: 33026818 DOI: 10.1152/ajplung.00381.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ozone is known to cause lung injury, and resident cells of the respiratory tract (i.e., epithelial cells and macrophages) respond to inhaled ozone in a variety of ways that affect their survival, morphology, and functioning. However, a complete understanding of the sex-associated and the cell type-specific gene expression changes in response to ozone exposure is still limited. Through transcriptome profiling, we aimed to analyze gene expression alterations and associated enrichment of biological pathways in three distinct cell type-enriched compartments of ozone-exposed murine lungs. We subchronically exposed adult male and female mice to 0.8 ppm ozone or filtered air. RNA-Seq was performed on airway epithelium-enriched airways, parenchyma, and purified airspace macrophages. Differential gene expression and biological pathway analyses were performed and supported by cellular and immunohistochemical analyses. While a majority of differentially expressed genes (DEGs) in ozone-exposed versus air-exposed groups were common between both sexes, sex-specific DEGs were also identified in all of the three tissue compartments. As compared with ozone-exposed males, ozone-exposed females had significant alterations in gene expression in three compartments. Pathways relevant to cell division and DNA repair were enriched in the ozone-exposed airways, indicating ozone-induced airway injury and repair, which was further supported by immunohistochemical analyses. In addition to cell division and DNA repair pathways, inflammatory pathways were also enriched within the parenchyma, supporting contribution by both epithelial and immune cells. Further, immune response and cytokine-cytokine receptor interactions were enriched in macrophages, indicating ozone-induced macrophage activation. Finally, our analyses also revealed the overall upregulation of mucoinflammation- and mucous cell metaplasia-associated pathways following ozone exposure.
Collapse
Affiliation(s)
- Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Thao Vo
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Kshitiz Paudel
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
76
|
IL-33-ST2 axis regulates myeloid cell differentiation and activation enabling effective club cell regeneration. Nat Commun 2020; 11:4786. [PMID: 32963227 PMCID: PMC7508874 DOI: 10.1038/s41467-020-18466-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/25/2020] [Indexed: 02/06/2023] Open
Abstract
Evidence points to an indispensable function of macrophages in tissue regeneration, yet the underlying molecular mechanisms remain elusive. Here we demonstrate a protective function for the IL-33-ST2 axis in bronchial epithelial repair, and implicate ST2 in myeloid cell differentiation. ST2 deficiency in mice leads to reduced lung myeloid cell infiltration, abnormal alternatively activated macrophage (AAM) function, and impaired epithelial repair post naphthalene-induced injury. Reconstitution of wild type (WT) AAMs to ST2-deficient mice completely restores bronchial re-epithelialization. Central to this mechanism is the direct effect of IL-33-ST2 signaling on monocyte/macrophage differentiation, self-renewal and repairing ability, as evidenced by the downregulation of key pathways regulating myeloid cell cycle, maturation and regenerative function of the epithelial niche in ST2−/− mice. Thus, the IL-33-ST2 axis controls epithelial niche regeneration by activating a large multi-cellular circuit, including monocyte differentiation into competent repairing AAMs, as well as group-2 innate lymphoid cell (ILC2)-mediated AAM activation. Signaling of IL-33 via its receptor, ST2, has been implicated in macrophage function in tissue repair. Here the authors show, using genetic mouse models and single-cell transcriptomic data, that the IL-33/ST2 axis regulates both ILC2-derived IL-13 and macrophage differentiation/reparative function required for club cell regeneration.
Collapse
|
77
|
Fourier N, Zolty M, Azriel A, Tedesco D, Levi BZ. MafK Mediates Chromatin Remodeling to Silence IRF8 Expression in Non-immune Cells in a Cell Type-SpecificManner. J Mol Biol 2020; 432:4544-4560. [PMID: 32534063 DOI: 10.1016/j.jmb.2020.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 11/17/2022]
Abstract
The regulation of gene expression is a result of a complex interplay between chromatin remodeling, transcription factors, and signaling molecules. Cell differentiation is accompanied by chromatin remodeling of specific loci to permanently silence genes that are not essential for the differentiated cell activity. The molecular cues that recruit the chromatin remodeling machinery are not well characterized. IRF8 is an immune-cell specific transcription factor and its expression is augmented by interferon-γ. Therefore, it serves as a model gene to elucidate the molecular mechanisms governing its silencing in non-immune cells. Ahigh-throughput shRNA library screen in IRF8 expression-restrictive cells enabled the identification of MafK as modulator of IRF8 silencing, affecting chromatin architecture. ChIP-Seq analysis revealed three MafK binding regions (-25 kb, -20 kb, and IRF8 6th intron) within the IRF8 locus. These MafK binding sites are sufficient to repress a reporter gene when cloned in genome-integrated lentiviral reporter constructs in only expression-restrictive cells. Conversely, plasmid-based constructs do not demonstrate such repressive effect. These results highlight the role of these MafK binding sites in mediating repressed chromatin assembly. Finally, a more thorough genomic analysis was performed, using CRISPR-Cas9 to delete MafK-int6 binding region in IRF8 expression-restrictive cells. Deleted clones exhibited an accessible chromatin conformation within the IRF8 locus that was accompanied by a significant increase in basal expression of IRF8 that was further induced by interferon-γ. Taken together, we identified and characterized several MafK binding elements within the IRF8 locus that mediate repressive chromatin conformation resulting in the silencing of IRF8 expression in a celltype-specific manner.
Collapse
Affiliation(s)
- Nitsan Fourier
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Maya Zolty
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Aviva Azriel
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Ben-Zion Levi
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
78
|
Nasser H, Adhikary P, Abdel-Daim A, Noyori O, Panaampon J, Kariya R, Okada S, Ma W, Baba M, Takizawa H, Yamane M, Niwa H, Suzu S. Establishment of bone marrow-derived M-CSF receptor-dependent self-renewing macrophages. Cell Death Discov 2020; 6:63. [PMID: 32714570 PMCID: PMC7378060 DOI: 10.1038/s41420-020-00300-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Recent studies have revealed that tissue macrophages are derived from yolk sac precursors or fetal liver monocytes, in addition to bone marrow monocytes. The relative contribution of these cells to the tissue macrophage pool is not fully understood, but embryo-derived cells are supposed to be more important because of their capacity to self-renew. Here, we show the presence of adult bone marrow-derived macrophages that retain self-renewing capacity. The self-renewing macrophages were readily obtained by long-term culture of mouse bone marrow cells with macrophage colony-stimulating factor (M-CSF), a key cytokine for macrophage development. They were non-tumorigenic and proliferated in the presence of M-CSF in unlimited numbers. Despite several differences from non-proliferating macrophages, they retained many features of cells of the monocytic lineage, including the differentiation into dendritic cells or osteoclasts. Among the transcription factors involved in the self-renewal of embryonic stem cells, Krüppel-like factor 2 (KLF2) was strongly upregulated upon M-CSF stimulation in the self-renewing macrophages, which was accompanied by the downregulation of MafB, a transcription factor that suppresses KLF2 expression. Indeed, knockdown of KLF2 led to cell cycle arrest and diminished cell proliferation in the self-renewing macrophages. Our new cell model would be useful to unravel differences in phenotype, function, and molecular mechanism of proliferation among self-renewing macrophages with different origins.
Collapse
Affiliation(s)
- Hesham Nasser
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811 Japan
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811 Japan
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, 41511 Egypt
| | - Partho Adhikary
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811 Japan
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811 Japan
- Present Address: Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, V6T 1Z3 Canada
| | - Amira Abdel-Daim
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811 Japan
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811 Japan
| | - Osamu Noyori
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811 Japan
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811 Japan
| | - Jutatip Panaampon
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811 Japan
| | - Ryusho Kariya
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811 Japan
| | - Seiji Okada
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811 Japan
| | - Wenjuan Ma
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811 Japan
| | - Masaya Baba
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811 Japan
| | - Hitoshi Takizawa
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811 Japan
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, 860-8556 Japan
| | - Mariko Yamane
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811 Japan
- Present Address: Laboratory for Bioinformatics Research, RIKEN Center for Biosystems Dynamics Research, Hyogo, 650-0047 Japan
| | - Hitoshi Niwa
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811 Japan
| | - Shinya Suzu
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0811 Japan
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, 860-0811 Japan
| |
Collapse
|
79
|
Lohrmann F, Forde AJ, Merck P, Henneke P. Control of myeloid cell density in barrier tissues. FEBS J 2020; 288:405-426. [PMID: 32502309 DOI: 10.1111/febs.15436] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/21/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022]
Abstract
The interface between the mammalian host and its environment is formed by barrier tissues, for example, of the skin, and the respiratory and the intestinal tracts. On the one hand, barrier tissues are colonized by site-adapted microbial communities, and on the other hand, they contain specific myeloid cell networks comprising macrophages, dendritic cells, and granulocytes. These immune cells are tightly regulated in function and cell number, indicating important roles in maintaining tissue homeostasis and immune balance in the presence of commensal microorganisms. The regulation of myeloid cell density and activation involves cell-autonomous 'single-loop circuits' including autocrine mechanisms. However, an array of microenvironmental factors originating from nonimmune cells and the microbiota, as well as the microanatomical structure, impose additional layers of regulation onto resident myeloid cells. This review discusses models integrating these factors into cell-specific programs to instruct differentiation and proliferation best suited for the maintenance and renewal of immune homeostasis in the tissue-specific environment.
Collapse
Affiliation(s)
- Florens Lohrmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Germany.,Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany.,Spemann Graduate School for Biology and Medicine, University of Freiburg, Germany.,IMM-PACT Clinician Scientist Program, Faculty of Medicine, University of Freiburg, Germany
| | - Aaron J Forde
- Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany.,Faculty of Biology, university of Freiburg, Germany
| | - Philipp Merck
- Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany
| | - Philipp Henneke
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Germany.,Institute for Immunodeficiency (IFI), Faculty of Medicine, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Germany
| |
Collapse
|
80
|
Giurisato E, Lonardi S, Telfer B, Lussoso S, Risa-Ebrí B, Zhang J, Russo I, Wang J, Santucci A, Finegan KG, Gray NS, Vermi W, Tournier C. Extracellular-Regulated Protein Kinase 5-Mediated Control of p21 Expression Promotes Macrophage Proliferation Associated with Tumor Growth and Metastasis. Cancer Res 2020; 80:3319-3330. [PMID: 32561530 DOI: 10.1158/0008-5472.can-19-2416] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 04/07/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
The presence of immunosuppressive macrophages that become activated in the tumor microenvironment constitutes a major factor responsible for tumor growth and malignancy. In line with this knowledge, we report here that macrophage proliferation is a significant feature of advanced stages of cancer. Moreover, we have found that a high proportion of proliferating macrophages in human tumors express ERK5. ERK5 was required for supporting the proliferation of macrophages in tumor grafts in mice. Furthermore, myeloid ERK5 deficiency negatively impacted the proliferation of both resident and infiltrated macrophages in metastatic lung nodules. ERK5 maintained the capacity of macrophages to proliferate by suppressing p21 expression to halt their differentiation program. Collectively, these data provide insight into the mechanism underpinning macrophage proliferation to support malignant tumor development, thereby strengthening the value of ERK5-targeted therapies to restore antitumor immunity through the blockade of protumorigenic macrophage activation. SIGNIFICANCE: These findings offer a new rationale for anti-ERK5 therapy to improve cancer patient outcomes by blocking the proliferative activity of tumor macrophages.
Collapse
Affiliation(s)
- Emanuele Giurisato
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy. .,Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Silvia Lonardi
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Brian Telfer
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Sarah Lussoso
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Blanca Risa-Ebrí
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Jingwei Zhang
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Ilaria Russo
- School of Medicine, Keel University, Keel, United Kingdom.,Department of Medicine-Infectious Diseases, Washington University, Saint Louis, Missouri
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Annalisa Santucci
- Department of Biotechnology Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Katherine G Finegan
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - William Vermi
- Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy.,Department of Pathology and Immunology, Washington University, Saint Louis, Missouri
| | - Cathy Tournier
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
81
|
Zhang H, Xu A, Sun X, Yang Y, Zhang L, Bai H, Ben J, Zhu X, Li X, Yang Q, Wang Z, Wu W, Yang D, Zhang Y, Xu Y, Chen Q. Self-Maintenance of Cardiac Resident Reparative Macrophages Attenuates Doxorubicin-Induced Cardiomyopathy Through the SR-A1-c-Myc Axis. Circ Res 2020; 127:610-627. [PMID: 32466726 DOI: 10.1161/circresaha.119.316428] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
RATIONALE Doxorubicin-induced cardiomyopathy (DiCM) is a primary cause of heart failure and mortality in cancer patients, in which macrophage-orchestrated inflammation serves as an essential pathological mechanism. However, the specific roles of tissue-resident and monocyte-derived macrophages in DiCM remain poorly understood. OBJECTIVE Uncovering the origins, phenotypes, and functions of proliferative cardiac resident macrophages and mechanistic insights into the self-maintenance of cardiac macrophage during DiCM progression. METHODS AND RESULTS Mice were administrated with doxorubicin to induce cardiomyopathy. Dynamic changes of resident and monocyte-derived macrophages were examined by lineage tracing, parabiosis, and bone marrow transplantation. We found that the monocyte-derived macrophages primarily exhibited a proinflammatory phenotype that dominated the whole DiCM pathological process and impaired cardiac function. In contrast, cardiac resident macrophages were vulnerable to doxorubicin insult. The survived resident macrophages exhibited enhanced proliferation and conferred a reparative role. Global or myeloid specifically ablation of SR-A1 (class A1 scavenger receptor) inhibited proliferation of cardiac resident reparative macrophages and, therefore, exacerbated cardiomyopathy in DiCM mice. Importantly, the detrimental effect of macrophage SR-A1 deficiency was confirmed by transplantation of bone marrow. At the mechanistic level, we show that c-Myc (Avian myelocytomatosis virus oncogene cellular homolog), a key transcriptional factor for the SR-A1-P38-SIRT1 (Sirtuin 1) pathway, mediated the effect of SR-A1 in reparative macrophage proliferation in DiCM. CONCLUSIONS The SR-A1-c-Myc axis may represent a promising target to treat DiCM through augmentation of cardiac resident reparative macrophage proliferation.
Collapse
MESH Headings
- Animals
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- Cardiomyopathy, Dilated/chemically induced
- Cardiomyopathy, Dilated/enzymology
- Cardiomyopathy, Dilated/pathology
- Cardiomyopathy, Dilated/prevention & control
- Cell Proliferation
- Cell Self Renewal
- Cells, Cultured
- Disease Models, Animal
- Doxorubicin
- Female
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- Macrophages/enzymology
- Macrophages/pathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardium/enzymology
- Myocardium/pathology
- Phenotype
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Scavenger Receptors, Class A/deficiency
- Scavenger Receptors, Class A/genetics
- Scavenger Receptors, Class A/metabolism
- Signal Transduction
- Ventricular Remodeling
Collapse
Affiliation(s)
- Hanwen Zhang
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Andi Xu
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Xuan Sun
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Department of Cardiology, Nanjing Drum Tower Hospital, China (X.S.)
| | - Yaqing Yang
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Lai Zhang
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Hui Bai
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Jingjing Ben
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Xudong Zhu
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Xiaoyu Li
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Qing Yang
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Zidun Wang
- Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, China (Z.W., D.Y.)
| | - Wei Wu
- Bioinformatics (W.W.), Nanjing Medical University, China
| | - Di Yang
- Department of Cardiology, First Affiliated Hospital with Nanjing Medical University, China (Z.W., D.Y.)
| | | | - Yong Xu
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| | - Qi Chen
- From the Department of Pathophysiology (H.Z., A.X., X.S., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.), Nanjing Medical University, China
- Key Laboratory of Targeted Intervention of Cardiovascular Diseases, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Jiangsu Province, China (H.Z., A.X., Y.Y., L.Z., H.B., J.B., X.Z., X.L., Q.Y., Y.X., Q.C.)
| |
Collapse
|
82
|
Yang HL, Wang CJ, Lai ZZ, Yang SL, Zheng ZM, Shi JW, Li MQ, Shao J. Decidual stromal cells maintain decidual macrophage homeostasis by secreting IL-24 in early pregnancy. Am J Reprod Immunol 2020; 84:e13261. [PMID: 32356306 DOI: 10.1111/aji.13261] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/10/2020] [Accepted: 04/27/2020] [Indexed: 12/23/2022] Open
Abstract
PROBLEM The state of self-renewal and self-maintain of decidual macrophages would be important for immune homeostasis at the maternal-fetal interface. The roles of interleukin (IL)-24 derived from decidual stromal cells (DSCs) on decidual macrophages have not been explored. METHOD OF STUDY IL-24 expression in DSCs was interfered by lentivirus, and the transcription levels of IL-24 in DSCs were verified by real time (RT)-PCR. The levels of IL-24 receptors were determined by flow cytometry assays. The effect of recombination human IL-24 (rhIL-24) on the differentiation and apoptosis of macrophages was analyzed by flow cytometry in vitro. The viability of macrophages was detected by Cell Counting Kit-8 assays. RESULTS The growth of DSCs was not affected obviously only by IL-24 knockdown while the growth of knockdown DSCs was inhibited significantly after co-cultured with decidual macrophages. The levels of IL-24 receptors (IL-20R1 and IL-22R1) were moderately to highly expressed on decidual macrophages and human macrophage cell line U937. The differentiation of decidual macrophages treated by rhIL-24 or co-cultured with IL-24 knockdown DSCs was not affected. Both apoptosis and viability of U937 cells were promoted by rhIL-24. The ratio of Bcl-2/Bax was down-regulated and Ki-67 was up-regulated by IL-24 treatment. The expression of Bcl-2/Bax was up-regulated while Ki-67 was down-regulated in U937 cells after co-cultured by IL-24 knockdown DSCs. CONCLUSION IL-24 secreted by DSCs promotes the renewal and homeostasis of decidual macrophages possibly via down-regulating the ratio of Bcl-2/Bax and up-regulating of the expression of Ki-67 in early pregnancy.
Collapse
Affiliation(s)
- Hui-Li Yang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Cheng-Jie Wang
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Zhen-Zhen Lai
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Shao-Liang Yang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Zi-Meng Zheng
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jia-Wei Shi
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Ming-Qing Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jun Shao
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
83
|
Bianchetto-Aguilera F, Tamassia N, Gasperini S, Calzetti F, Finotti G, Gardiman E, Montioli R, Bresciani D, Vermi W, Cassatella MA. Deciphering the fate of slan + -monocytes in human tonsils by gene expression profiling. FASEB J 2020; 34:9269-9284. [PMID: 32413173 DOI: 10.1096/fj.202000181r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/24/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022]
Abstract
Monocytic cells perform crucial homeostatic and defensive functions. However, their fate and characterization at the transcriptomic level in human tissues are partially understood, often as a consequence of the lack of specific markers allowing their unequivocal identification. The 6-sulfo LacNAc (slan) antigen identifies a subset of non-classical (NC) monocytes in the bloodstream, namely the slan+ -monocytes. In recent studies, we and other groups have reported that, in tonsils, slan marks dendritic cell (DC)-like cells, as defined by morphological, phenotypical, and functional criteria. However, subsequent investigations in lymphomas have uncovered a significant heterogeneity of tumor-infiltrating slan+ -cells, including a macrophage-like state. Based on their emerging role in tissue inflammation and cancer, herein we investigated slan+ -cell fate in tonsils by using a molecular-based approach. Hence, RNA from tonsil slan+ -cells, conventional CD1c+ DCs (cDC2) and CD11b+ CD14+ -macrophages was subjected to gene expression analysis. For comparison, transcriptomes were also obtained from blood cDC2, classical (CL), intermediate (INT), NC, and slan+ -monocytes. Data demonstrate that the main trajectory of human slan+ -monocytes infiltrating the tonsil tissue is toward a macrophage-like population, displaying molecular features distinct from those of tonsil CD11b+ CD14+ -macrophages and cDC2. These findings provide a novel view on the terminal differentiation path of slan+ -monocytes, which is relevant for inflammatory diseases and lymphomas.
Collapse
Affiliation(s)
| | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Sara Gasperini
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Federica Calzetti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Giulia Finotti
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Elisa Gardiman
- Department of Medicine, Section of General Pathology, University of Verona, Verona, Italy
| | - Riccardo Montioli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Debora Bresciani
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy
| | - William Vermi
- Department of Molecular and Translational Medicine, Section of Pathology, University of Brescia, Brescia, Italy.,Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
84
|
Lu J, Dalbeth N, Yin H, Li C, Merriman TR, Wei WH. Mouse models for human hyperuricaemia: a critical review. Nat Rev Rheumatol 2020; 15:413-426. [PMID: 31118497 DOI: 10.1038/s41584-019-0222-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hyperuricaemia (increased serum urate concentration) occurs mainly in higher primates, including in humans, because of inactivation of the gene encoding uricase during primate evolution. Individuals with hyperuricaemia might develop gout - a painful inflammatory arthritis caused by monosodium urate crystal deposition in articular structures. Hyperuricaemia is also associated with common chronic diseases, including hypertension, chronic kidney disease, type 2 diabetes and cardiovascular disease. Many mouse models have been developed to investigate the causal mechanisms for hyperuricaemia. These models are highly diverse and can be divided into two broad categories: mice with genetic modifications (genetically induced models) and mice exposed to certain environmental factors (environmentally induced models; for example, pharmaceutical or dietary induction). This Review provides an overview of the mouse models of hyperuricaemia and the relevance of these models to human hyperuricaemia, with an emphasis on those models generated through genetic modifications. The challenges in developing and comparing mouse models of hyperuricaemia and future research directions are also outlined.
Collapse
Affiliation(s)
- Jie Lu
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand.,Shandong Provincial Key Laboratory of Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, the Affiliated Hospital of Qingdao University, Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Nicola Dalbeth
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Huiyong Yin
- Chinese Academy of Sciences (CAS) Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences (SIBS), CAS, Shanghai, China
| | - Changgui Li
- Shandong Provincial Key Laboratory of Metabolic Diseases, Department of Endocrinology and Metabolic Diseases, the Affiliated Hospital of Qingdao University, Institute of Metabolic Diseases, Qingdao University, Qingdao, China
| | - Tony R Merriman
- Department of Biochemistry, University of Otago, Dunedin, New Zealand.
| | - Wen-Hua Wei
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
85
|
Rosas-Ballina M, Guan XL, Schmidt A, Bumann D. Classical Activation of Macrophages Leads to Lipid Droplet Formation Without de novo Fatty Acid Synthesis. Front Immunol 2020; 11:131. [PMID: 32132994 PMCID: PMC7040478 DOI: 10.3389/fimmu.2020.00131] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 01/17/2020] [Indexed: 01/17/2023] Open
Abstract
Altered lipid metabolism in macrophages is associated with various important inflammatory conditions. Although lipid metabolism is an important target for therapeutic intervention, the metabolic requirement involved in lipid accumulation during pro-inflammatory activation of macrophages remains incompletely characterized. We show here that macrophage activation with IFNγ results in increased aerobic glycolysis, iNOS-dependent inhibition of respiration, and accumulation of triacylglycerol. Surprisingly, metabolite tracing with 13C-labeled glucose revealed that the glucose contributed to the glycerol groups in triacylglycerol (TAG), rather than to de novo synthesis of fatty acids. This is in stark contrast to the otherwise similar metabolism of cancer cells, and previous results obtained in activated macrophages and dendritic cells. Our results establish a novel metabolic pathway whereby glucose provides glycerol to the headgroup of TAG during classical macrophage activation.
Collapse
Affiliation(s)
| | - Xue Li Guan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Dirk Bumann
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
86
|
Wheeler MA, Clark IC, Tjon EC, Li Z, Zandee SEJ, Couturier CP, Watson BR, Scalisi G, Alkwai S, Rothhammer V, Rotem A, Heyman JA, Thaploo S, Sanmarco LM, Ragoussis J, Weitz DA, Petrecca K, Moffitt JR, Becher B, Antel JP, Prat A, Quintana FJ. MAFG-driven astrocytes promote CNS inflammation. Nature 2020; 578:593-599. [PMID: 32051591 PMCID: PMC8049843 DOI: 10.1038/s41586-020-1999-0] [Citation(s) in RCA: 312] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 12/16/2019] [Indexed: 01/14/2023]
Abstract
Multiple sclerosis is a chronic inflammatory disease of the CNS1. Astrocytes contribute to the pathogenesis of multiple sclerosis2, but little is known about the heterogeneity of astrocytes and its regulation. Here we report the analysis of astrocytes in multiple sclerosis and its preclinical model experimental autoimmune encephalomyelitis (EAE) by single-cell RNA sequencing in combination with cell-specific Ribotag RNA profiling, assay for transposase-accessible chromatin with sequencing (ATAC-seq), chromatin immunoprecipitation with sequencing (ChIP-seq), genome-wide analysis of DNA methylation and in vivo CRISPR-Cas9-based genetic perturbations. We identified astrocytes in EAE and multiple sclerosis that were characterized by decreased expression of NRF2 and increased expression of MAFG, which cooperates with MAT2α to promote DNA methylation and represses antioxidant and anti-inflammatory transcriptional programs. Granulocyte-macrophage colony-stimulating factor (GM-CSF) signalling in astrocytes drives the expression of MAFG and MAT2α and pro-inflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, multiple sclerosis. Our results identify candidate therapeutic targets in multiple sclerosis.
Collapse
Affiliation(s)
- Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Iain C Clark
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily C Tjon
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhaorong Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephanie E J Zandee
- Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Charles P Couturier
- Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Brianna R Watson
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Giulia Scalisi
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Alkwai
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA
- Centre of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Assaf Rotem
- Department of Physics, Harvard University, Cambridge, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - John A Heyman
- Department of Physics, Harvard University, Cambridge, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Shravan Thaploo
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Liliana M Sanmarco
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiannis Ragoussis
- McGill University and Genome Quebec Innovation Centre, Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - David A Weitz
- Department of Physics, Harvard University, Cambridge, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Kevin Petrecca
- Montreal Neurological Institute and Hospital, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Jeffrey R Moffitt
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Alexandre Prat
- Neuroimmunology Unit, Centre de recherche du CHUM (CRCHUM), Montreal, Quebec, Canada
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Centre of Excellence for Biomedicine, Brigham and Women's Hospital, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
87
|
Dong Y, Arif AA, Guo J, Ha Z, Lee-Sayer SSM, Poon GFT, Dosanjh M, Roskelley CD, Huan T, Johnson P. CD44 Loss Disrupts Lung Lipid Surfactant Homeostasis and Exacerbates Oxidized Lipid-Induced Lung Inflammation. Front Immunol 2020; 11:29. [PMID: 32082314 PMCID: PMC7002364 DOI: 10.3389/fimmu.2020.00029] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/08/2020] [Indexed: 12/26/2022] Open
Abstract
Alveolar macrophages (AMs) are CD44 expressing cells that reside in the alveolar space where they maintain lung homeostasis by serving critical roles in immunosurveillance and lipid surfactant catabolism. AMs lacking CD44 are unable to bind the glycosaminoglycan, hyaluronan, which compromises their survival and leads to reduced numbers of AMs in the lung. Using RNA sequencing, lipidomics and multiparameter flow cytometry, we demonstrate that CD44−/− mice have impaired AM lipid homeostasis and increased surfactant lipids in the lung. CD44−/− AMs had increased expression of CD36, a lipid scavenger receptor, as well as increased intracellular lipid droplets, giving them a foamy appearance. RNA sequencing revealed the differential expression of genes associated with lipid efflux and metabolism in CD44−/− AMs. Lipidomic analysis showed increased lipids in both the supernatant and cell pellet extracted from the bronchoalveolar lavage of CD44−/− mice. Phosphatidylcholine species, cholesterol, oxidized phospholipids and levels of reactive oxygen species (ROS) were increased in CD44−/− AMs. Oxidized phospholipids were more cytotoxic to CD44−/− AMs and induced greater lung inflammation in CD44−/− mice. Reconstitution of CD44+/+ mice with CD44−/− bone marrow as well as adoptive transfer of CD44−/− AMs into CD44+/+ mice showed that lipid accumulation in CD44−/− AMs occurred irrespective of the lung environment, suggesting a cell intrinsic defect. Administration of colony stimulating factor 2 (CSF-2), a critical factor in AM development and maintenance, increased AM numbers in CD44−/− mice and decreased phosphatidylcholine levels in the bronchoalveolar lavage, but was unable to decrease intracellular lipid accumulation in CD44−/− AMs. Peroxisome proliferator-activated receptor gamma (PPARγ), downstream of CSF-2 signaling and a regulator of lipid metabolism, was reduced in the nucleus of CD44−/− AMs, and PPARγ inhibition in normal AMs increased their lipid droplets. Thus, CD44 deficiency causes defects in AMs that lead to abnormal lipid accumulation and oxidation, which exacerbates oxidized lipid-induced lung inflammation. Collectively, these findings implicate CD44 as a regulator of lung homeostasis and inflammation.
Collapse
Affiliation(s)
- Yifei Dong
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Arif A Arif
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Jian Guo
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | - Zongyi Ha
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Sally S M Lee-Sayer
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Grace F T Poon
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Manisha Dosanjh
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| | - Calvin D Roskelley
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Tao Huan
- Department of Chemistry, University of British Columbia, Vancouver, BC, Canada
| | - Pauline Johnson
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
88
|
Li F, Okreglicka KM, Pohlmeier LM, Schneider C, Kopf M. Fetal monocytes possess increased metabolic capacity and replace primitive macrophages in tissue macrophage development. EMBO J 2020; 39:e103205. [PMID: 31894879 DOI: 10.15252/embj.2019103205] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/20/2019] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
Tissue-resident macrophages (MΦTR ) originate from at least two distinct waves of erythro-myeloid progenitors (EMP) arising in the yolk sac (YS) at E7.5 and E8.5 with the latter going through a liver monocyte intermediate. The relative potential of these precursors in determining development and functional capacity of MΦTR remains unclear. Here, we studied development of alveolar macrophages (AM) after single and competitive transplantation of different precursors from YS, fetal liver, and fetal lung into neonatal Csf2ra-/- mice, which lack endogenous AM. Fetal monocytes, promoted by Myb, outcompeted primitive MΦ (pMΦ) in empty AM niches and preferentially developed to mature AM, which is associated with enhanced mitochondrial respiratory and glycolytic capacity and repression of the transcription factors c-Maf and MafB. Interestingly, AM derived from pMΦ failed to efficiently clear alveolar proteinosis and protect from fatal lung failure following influenza virus infection. Thus, our data demonstrate superior developmental and functional capacity of fetal monocytes over pMΦ in AM development and underlying mechanisms explaining replacement of pMΦ in fetal tissues.
Collapse
Affiliation(s)
- Fengqi Li
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | | | - Lea Maria Pohlmeier
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| | - Christoph Schneider
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland.,Institute of Physiology, University of Zürich, Zürich, Switzerland
| | - Manfred Kopf
- Department of Biology, Institute of Molecular Health Sciences, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
89
|
Evren E, Ringqvist E, Willinger T. Origin and ontogeny of lung macrophages: from mice to humans. Immunology 2019; 160:126-138. [PMID: 31715003 DOI: 10.1111/imm.13154] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/19/2022] Open
Abstract
Macrophages are tissue-resident myeloid cells with essential roles in host defense, tissue repair, and organ homeostasis. The lung harbors a large number of macrophages that reside in alveoli. As a result of their strategic location, alveolar macrophages are critical sentinels of healthy lung function and barrier immunity. They phagocytose inhaled material and initiate protective immune responses to pathogens, while preventing excessive inflammatory responses and tissue damage. Apart from alveolar macrophages, other macrophage populations are found in the lung and recent single-cell RNA-sequencing studies indicate that lung macrophage heterogeneity is greater than previously appreciated. The cellular origin and development of mouse lung macrophages has been extensively studied, but little is known about the ontogeny of their human counterparts, despite the importance of macrophages for lung health. In this context, humanized mice (mice with a human immune system) can give new insights into the biology of human lung macrophages by allowing in vivo studies that are not possible in humans. In particular, we have created humanized mouse models that support the development of human lung macrophages in vivo. In this review, we will discuss the heterogeneity, development, and homeostasis of lung macrophages. Moreover, we will highlight the impact of age, the microbiota, and pathogen exposure on lung macrophage function. Altered macrophage function has been implicated in respiratory infections as well as in common allergic and inflammatory lung diseases. Therefore, understanding the functional heterogeneity and ontogeny of lung macrophages should help to develop future macrophage-based therapies for important lung diseases in humans.
Collapse
Affiliation(s)
- Elza Evren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Emma Ringqvist
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tim Willinger
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
90
|
Hu W, Yang S, Shimada Y, Münch M, Marín-Juez R, Meijer AH, Spaink HP. Infection and RNA-seq analysis of a zebrafish tlr2 mutant shows a broad function of this toll-like receptor in transcriptional and metabolic control and defense to Mycobacterium marinum infection. BMC Genomics 2019; 20:878. [PMID: 31747871 PMCID: PMC6869251 DOI: 10.1186/s12864-019-6265-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background The function of Toll-like receptor 2 (TLR2) in host defense against pathogens, especially Mycobacterium tuberculosis (Mtb) is poorly understood. To investigate the role of TLR2 during mycobacterial infection, we analyzed the response of tlr2 zebrafish mutant larvae to infection with Mycobacterium marinum (Mm), a close relative to Mtb, as a model for tuberculosis. We measured infection phenotypes and transcriptome responses using RNA deep sequencing in mutant and control larvae. Results tlr2 mutant embryos at 2 dpf do not show differences in numbers of macrophages and neutrophils compared to control embryos. However, we found substantial changes in gene expression in these mutants, particularly in metabolic pathways, when compared with the heterozygote tlr2+/− control. At 4 days after Mm infection, the total bacterial burden and the presence of extracellular bacteria were higher in tlr2−/− larvae than in tlr2+/−, or tlr2+/+ larvae, whereas granuloma numbers were reduced, showing a function of Tlr2 in zebrafish host defense. RNAseq analysis of infected tlr2−/− versus tlr2+/− shows that the number of up-regulated and down-regulated genes in response to infection was greatly diminished in tlr2 mutants by at least 2 fold and 10 fold, respectively. Analysis of the transcriptome data and qPCR validation shows that Mm infection of tlr2 mutants leads to decreased mRNA levels of genes involved in inflammation and immune responses, including il1b, tnfb, cxcl11aa/ac, fosl1a, and cebpb. Furthermore, RNAseq analyses revealed that the expression of genes for Maf family transcription factors, vitamin D receptors, and Dicps proteins is altered in tlr2 mutants with or without infection. In addition, the data indicate a function of Tlr2 in the control of induction of cytokines and chemokines, such as the CXCR3-CXCL11 signaling axis. Conclusion The transcriptome and infection burden analyses show a function of Tlr2 as a protective factor against mycobacteria. Transcriptome analysis revealed tlr2-specific pathways involved in Mm infection, which are related to responses to Mtb infection in human macrophages. Considering its dominant function in control of transcriptional processes that govern defense responses and metabolism, the TLR2 protein can be expected to be also of importance for other infectious diseases and interactions with the microbiome.
Collapse
Affiliation(s)
- Wanbin Hu
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands
| | - Shuxin Yang
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yasuhito Shimada
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Magnus Münch
- Mathematical Institute, Leiden University, Leiden, the Netherlands.,Department of Epidemiology & Biostatistics, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Rubén Marín-Juez
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.,Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, Germany
| | - Annemarie H Meijer
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands
| | - Herman P Spaink
- Institute of Biology, Leiden University, P.O. Box 9505, 2300 RA, Leiden, the Netherlands.
| |
Collapse
|
91
|
Personnic N, Striednig B, Lezan E, Manske C, Welin A, Schmidt A, Hilbi H. Quorum sensing modulates the formation of virulent Legionella persisters within infected cells. Nat Commun 2019; 10:5216. [PMID: 31740681 PMCID: PMC6861284 DOI: 10.1038/s41467-019-13021-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 10/14/2019] [Indexed: 12/21/2022] Open
Abstract
The facultative intracellular bacterium Legionella pneumophila replicates in environmental amoebae and in lung macrophages, and causes Legionnaires' disease. Here we show that L. pneumophila reversibly forms replicating and nonreplicating subpopulations of similar size within amoebae. The nonreplicating bacteria are viable and metabolically active, display increased antibiotic tolerance and a distinct proteome, and show high virulence as well as the capacity to form a degradation-resistant compartment. Upon infection of naïve or interferon-γ-activated macrophages, the nonreplicating subpopulation comprises ca. 10% or 50%, respectively, of the total intracellular bacteria; hence, the nonreplicating subpopulation is of similar size in amoebae and activated macrophages. The numbers of nonreplicating bacteria within amoebae are reduced in the absence of the autoinducer synthase LqsA or other components of the Lqs quorum-sensing system. Our results indicate that virulent, antibiotic-tolerant subpopulations of L. pneumophila are formed during infection of evolutionarily distant phagocytes, in a process controlled by the Lqs system.
Collapse
Affiliation(s)
- Nicolas Personnic
- Institute for Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland.
| | - Bianca Striednig
- Institute for Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland
| | - Emmanuelle Lezan
- Proteomics Core Facility, Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| | - Christian Manske
- Max von Pettenkofer Institute, Ludwig-Maximilians University Munich, Pettenkoferstrasse 9a, 80336, Munich, Germany
| | - Amanda Welin
- Institute for Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Klingelbergstrasse 50/70, 4056, Basel, Switzerland
| | - Hubert Hilbi
- Institute for Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland
| |
Collapse
|
92
|
Kishimoto I, Okano T, Nishimura K, Motohashi T, Omori K. Early Development of Resident Macrophages in the Mouse Cochlea Depends on Yolk Sac Hematopoiesis. Front Neurol 2019; 10:1115. [PMID: 31695671 PMCID: PMC6817595 DOI: 10.3389/fneur.2019.01115] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/07/2019] [Indexed: 12/20/2022] Open
Abstract
Resident macrophages reside in all tissues throughout the body and play a central role in both tissue homeostasis and inflammation. Although the inner ear was once believed to be "immune-privileged," recent studies have shown that macrophages are distributed in the cochlea and may play important roles in the immune system thereof. Resident macrophages have heterogeneous origins among tissues and throughout developmental stages. However, the origins of embryonic cochlear macrophages remain unknown. Here, we show that the early development of resident macrophages in the mouse cochlea depends on yolk sac hematopoiesis. Accordingly, our results found that macrophages emerging around the developing otocyst at E10.5 exhibited dynamic changes in distribution and in situ proliferative capacity during embryonic and neonatal stages. Cochlear examination in Csf1r-null mice revealed a substantial decrease in the number of Iba1-positive macrophages in the spiral ganglion and spiral ligament, whereas they were still observed in the cochlear mesenchyme or on the intraluminal surface of the perilymphatic space. Our results demonstrated that two subtypes of resident macrophages are present in the embryonic cochlea, one being Csf1r-dependent macrophages that originate from the yolk sac and the other being Csf1r-independent macrophages that appear to be derived from the fetal liver via systemic circulation. We consider the present study to be a starting point for elucidating the roles of embryonic cochlear resident macrophages. Furthermore, resident macrophages in the embryonic cochlea could be a novel target for the treatment of various inner ear disorders.
Collapse
Affiliation(s)
- Ippei Kishimoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takayuki Okano
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Koji Nishimura
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tsutomu Motohashi
- Department of Tissue and Organ Development, Regeneration, and Advanced Medical Science, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Koichi Omori
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
93
|
Abstract
The transcription factor MafB regulates macrophage differentiation. However, studies on
the phenotype of Mafb-deficient macrophages are still limited. Recently,
it was shown that the specific expression of MafB permits macrophages to be distinguished
from dendritic cells. In addition, MafB has been reported to be involved in various
diseases related to macrophages. Studies using macrophage-specific
Mafb-deficient mice show that MafB is linked to atherosclerosis,
autoimmunity, obesity, and ischemic stroke, all of which exhibit macrophage abnormality.
Therefore, MafB is hypothesized to be indispensable for the regulation of macrophages to
maintain systemic homeostasis and may serve as an innovative target for treating
macrophage-related diseases.
Collapse
Affiliation(s)
- Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Laboratory Animal Resource Center (LARC), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Tsunakawa
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Hyojung Jeon
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Laboratory Animal Resource Center (LARC), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Manoj Kumar Yadav
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan.,Laboratory Animal Resource Center (LARC), Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
94
|
Wen Y, Lu X, Ren J, Privratsky JR, Yang B, Rudemiller NP, Zhang J, Griffiths R, Jain MK, Nedospasov SA, Liu BC, Crowley SD. KLF4 in Macrophages Attenuates TNF α-Mediated Kidney Injury and Fibrosis. J Am Soc Nephrol 2019; 30:1925-1938. [PMID: 31337692 PMCID: PMC6779357 DOI: 10.1681/asn.2019020111] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Polarized macrophage populations can orchestrate both inflammation of the kidney and tissue repair during CKD. Proinflammatory M1 macrophages initiate kidney injury, but mechanisms through which persistent M1-dependent kidney damage culminates in fibrosis require elucidation. Krüppel-like factor 4 (KLF4), a zinc-finger transcription factor that suppresses inflammatory signals, is an essential regulator of macrophage polarization in adipose tissues, but the effect of myeloid KLF4 on CKD progression is unknown. METHODS We used conditional mutant mice lacking KLF4 or TNFα (KLF4's downstream effector) selectively in myeloid cells to investigate macrophage KLF4's role in modulating CKD progression in two models of CKD that feature robust macrophage accumulation, nephrotoxic serum nephritis, and unilateral ureteral obstruction. RESULTS In these murine CKD models, KLF4 deficiency in macrophages infiltrating the kidney augmented their M1 polarization and exacerbated glomerular matrix deposition and tubular epithelial damage. During the induced injury in these models, macrophage-specific KLF4 deletion also exacerbated kidney fibrosis, with increased levels of collagen 1 and α-smooth muscle actin in the injured kidney. CD11b+Ly6Chi myeloid cells isolated from injured kidneys expressed higher levels of TNFα mRNA versus wild-type controls. In turn, mice bearing macrophage-specific deletion of TNFα exhibited decreased glomerular and tubular damage and attenuated kidney fibrosis in the models. Moreover, treatment with the TNF receptor-1 inhibitor R-7050 during nephrotoxic serum nephritis reduced damage, fibrosis, and necroptosis in wild-type mice and mice with KLF4-deficient macrophages, and abrogated the differences between the two groups in these parameters. CONCLUSIONS These data indicate that macrophage KLF4 ameliorates CKD by mitigating TNF-dependent injury and fibrosis.
Collapse
Affiliation(s)
- Yi Wen
- Division of Nephrology
- Departments of Medicine and
| | - Xiaohan Lu
- Division of Nephrology
- Departments of Medicine and
| | - Jiafa Ren
- Division of Nephrology
- Departments of Medicine and
| | - Jamie R Privratsky
- Anesthesiology, Durham VA and Duke University Medical Center, Durham, North Carolina
| | - Bo Yang
- Division of Nephrology
- Departments of Medicine and
| | | | | | | | - Mukesh K Jain
- Department of Medicine, Case Cardiovascular Research Institute, Harrington Heart and Vascular Institute, University Hospitals Case Medical Center, Cleveland, Ohio
| | - Sergei A Nedospasov
- Laboratory of Molecular Immunology, Engelhardt Institute of Molecular Biology, Lomonosov Moscow State University, Moscow, Russia; and
| | - Bi Cheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University, Nanjing, China
| | | |
Collapse
|
95
|
Gualdrini F, Natoli G. Transcriptional repressors as guardians of tissue macrophage identity. EMBO J 2019; 38:e103271. [PMID: 31529710 DOI: 10.15252/embj.2019103271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The association between specific transcription factors (TFs) and defined tissue-specific macrophage phenotypes is far from being univocal. Many TFs that have been associated with tissue-specific macrophages have relatively broad expression profiles suggesting the critical involvement of combinatorial regulation by multiple TFs in bringing about specific phenotypes. In the current issue of The EMBO Journal, Rauschmeier et al (2019) report the identification of the transcriptional repressors BHLHE40 and BHLHE41 as novel regulators of tissue-specific macrophage properties that work both to promote alveolar macrophage (AM) identity and to suppress alternative tissue-specific phenotypes.
Collapse
Affiliation(s)
| | - Gioacchino Natoli
- Humanitas University, Pieve Emanuele, Milano, Italy.,Humanitas Clinical Research Institute, IRCCS, Rozzano, Milano, Italy
| |
Collapse
|
96
|
Okabe Y. Molecular control of the identity of tissue-resident macrophages. Int Immunol 2019; 30:485-491. [PMID: 30371831 DOI: 10.1093/intimm/dxy019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/21/2018] [Indexed: 12/23/2022] Open
Abstract
Macrophages are present in virtually almost all tissues, exhibiting highly heterogeneous phenotypes as a consequence of adaptation to local tissue environments. Tissue-resident macrophages perform specialized functions that are essential for the maintenance of tissue homeostasis, and abnormalities of their functions are linked to various pathologies. Recent advances have shown that tissue-specific transcriptional programs are responsible for functional specialization of macrophages in different tissues. Here, I discuss the molecular basis of tissue-resident macrophage specialization and how it is regulated by tissue environmental cues.
Collapse
Affiliation(s)
- Yasutaka Okabe
- Laboratory of Molecular Genetics, Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| |
Collapse
|
97
|
Rauschmeier R, Gustafsson C, Reinhardt A, A-Gonzalez N, Tortola L, Cansever D, Subramanian S, Taneja R, Rossner MJ, Sieweke MH, Greter M, Månsson R, Busslinger M, Kreslavsky T. Bhlhe40 and Bhlhe41 transcription factors regulate alveolar macrophage self-renewal and identity. EMBO J 2019; 38:e101233. [PMID: 31414712 DOI: 10.15252/embj.2018101233] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 07/18/2019] [Accepted: 07/30/2019] [Indexed: 12/23/2022] Open
Abstract
Tissues in multicellular organisms are populated by resident macrophages, which perform both generic and tissue-specific functions. The latter are induced by signals from the microenvironment and rely on unique tissue-specific molecular programs requiring the combinatorial action of tissue-specific and broadly expressed transcriptional regulators. Here, we identify the transcription factors Bhlhe40 and Bhlhe41 as novel regulators of alveolar macrophages (AMs)-a population that provides the first line of immune defense and executes homeostatic functions in lung alveoli. In the absence of these factors, AMs exhibited decreased proliferation that resulted in a severe disadvantage of knockout AMs in a competitive setting. Gene expression analyses revealed a broad cell-intrinsic footprint of Bhlhe40/Bhlhe41 deficiency manifested by a downregulation of AM signature genes and induction of signature genes of other macrophage lineages. Genome-wide characterization of Bhlhe40 DNA binding suggested that these transcription factors directly repress the expression of lineage-inappropriate genes in AMs. Taken together, these results identify Bhlhe40 and Bhlhe41 as key regulators of AM self-renewal and guardians of their identity.
Collapse
Affiliation(s)
- René Rauschmeier
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Charlotte Gustafsson
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Annika Reinhardt
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Noelia A-Gonzalez
- Institute of Immunology, University of Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Münster, Münster, Germany
| | - Luigi Tortola
- Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Dilay Cansever
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sethuraman Subramanian
- CNRS, INSERM, CIML, Aix Marseille University, Marseille, France.,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), Berlin, Germany.,Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Reshma Taneja
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | - Moritz J Rossner
- Department of Psychiatry, Molecular Neurobiology, Ludwig Maximilian University, Munich, Germany
| | - Michael H Sieweke
- CNRS, INSERM, CIML, Aix Marseille University, Marseille, France.,Max-Delbrück-Centrum für Molekulare Medizin in der Helmholtzgemeinschaft (MDC), Berlin, Germany.,Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Robert Månsson
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden.,Hematology Center, Karolinska University Hospital, Stockholm, Sweden
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Taras Kreslavsky
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria.,Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
98
|
Locally renewing resident synovial macrophages provide a protective barrier for the joint. Nature 2019; 572:670-675. [PMID: 31391580 DOI: 10.1038/s41586-019-1471-1] [Citation(s) in RCA: 387] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 07/05/2019] [Indexed: 12/13/2022]
Abstract
Macrophages are considered to contribute to chronic inflammatory diseases such as rheumatoid arthritis1. However, both the exact origin and the role of macrophages in inflammatory joint disease remain unclear. Here we use fate-mapping approaches in conjunction with three-dimensional light-sheet fluorescence microscopy and single-cell RNA sequencing to perform a comprehensive spatiotemporal analysis of the composition, origin and differentiation of subsets of macrophages within healthy and inflamed joints, and study the roles of these macrophages during arthritis. We find that dynamic membrane-like structures, consisting of a distinct population of CX3CR1+ tissue-resident macrophages, form an internal immunological barrier at the synovial lining and physically seclude the joint. These barrier-forming macrophages display features that are otherwise typical of epithelial cells, and maintain their numbers through a pool of locally proliferating CX3CR1- mononuclear cells that are embedded into the synovial tissue. Unlike recruited monocyte-derived macrophages, which actively contribute to joint inflammation, these epithelial-like CX3CR1+ lining macrophages restrict the inflammatory reaction by providing a tight-junction-mediated shield for intra-articular structures. Our data reveal an unexpected functional diversification among synovial macrophages and have important implications for the general role of macrophages in health and disease.
Collapse
|
99
|
Hu G, Dong T, Wang S, Jing H, Chen J. Vitamin D 3-vitamin D receptor axis suppresses pulmonary emphysema by maintaining alveolar macrophage homeostasis and function. EBioMedicine 2019; 45:563-577. [PMID: 31278070 PMCID: PMC6642288 DOI: 10.1016/j.ebiom.2019.06.039] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 02/06/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is characterized by emphysema and/or obstructive bronchiolitis. Deficiency in vitamin D3 (VD3), which regulates gene expression through binding to vitamin D receptor (VDR), is associated with high risks of COPD susceptibility. Alveolar macrophages (AM), which are generated during early ontogeny and maintained in alveoli by self-renewal in response to cytokine GM-CSF, are positively correlated with severity of emphysema. However, whether and how VD3, VDR and AM interact to contribute to COPD pathogenesis at the molecular and cellular levels are largely unknown. Methods We used systems biology approaches to analyze gene expression in mouse macrophages from different tissues to identify key transcription factors (TF) for AM and infer COPD disease genes. We used RNA-seq and ChIP-seq to identify genes that are regulated by VD3 in AM. We used VDR-deficient (Vdr−/−) mice to investigate the role of VD3-VDR axis in the pathogenesis of COPD and characterized the transcriptional and functional alterations of Vdr−/− AM. Findings We find that VDR is a key TF for AM and a COPD disease gene. VDR is highly expressed in AM and in response to VD3 inhibits GM-CSF-induced AM proliferation. In Vdr−/− AM, genes involved in proliferation and immune response are upregulated. Consistently, Vdr−/− mice progressively accumulate AM and concomitantly develop emphysema without apparent infiltration of immune cells into the lung tissue. Intratracheal transfer of Vdr−/− AM into wildtype mice readily induces emphysema. The production of reactive oxygen species at basal level and in response to heme or lipopolysaccharide is elevated in Vdr−/− AM and suppressed by VD3 in wildtype AM. Interpretation These results show that the VD3-VDR axis is critical to counteract GM-CSF-induced AM proliferation and defect in this regulation leads to altered AM homeostasis and function. Our findings identify that VD3 deficiency contributes to emphysema by altering AM function without contributing to bronchiolitis. Our findings also suggest possibilities of modulating the VD3-VDR axis for inhibiting emphysema in COPD patients.
Collapse
Affiliation(s)
- Guangan Hu
- David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Ting Dong
- David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Sisi Wang
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, China
| | - Hongyu Jing
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun 130021, China
| | - Jianzhu Chen
- David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
100
|
Singh T, Colberg JK, Sarmiento L, Chaves P, Hansen L, Bsharat S, Cataldo LR, Dudenhöffer-Pfeifer M, Fex M, Bryder D, Holmberg D, Sitnicka E, Cilio C, Prasad RB, Artner I. Loss of MafA and MafB expression promotes islet inflammation. Sci Rep 2019; 9:9074. [PMID: 31235823 PMCID: PMC6591483 DOI: 10.1038/s41598-019-45528-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 06/06/2019] [Indexed: 12/15/2022] Open
Abstract
Maf transcription factors are critical regulators of beta-cell function. We have previously shown that reduced MafA expression in human and mouse islets is associated with a pro-inflammatory gene signature. Here, we investigate if the loss of Maf transcription factors induced autoimmune processes in the pancreas. Transcriptomics analysis showed expression of pro-inflammatory as well as immune cell marker genes. However, clusters of CD4+ T and B220+ B cells were associated primarily with adult MafA−/−MafB+/−, but not MafA−/− islets. MafA expression was detected in the thymus, lymph nodes and bone marrow suggesting a novel role of MafA in regulating immune-cell function. Analysis of pancreatic lymph node cells showed activation of CD4+ T cells, but lack of CD8+ T cell activation which also coincided with an enrichment of naïve CD8+ T cells. Further analysis of T cell marker genes revealed a reduction of T cell receptor signaling gene expression in CD8, but not in CD4+ T cells, which was accompanied with a defect in early T cell receptor signaling in mutant CD8+ T cells. These results suggest that loss of MafA impairs both beta- and T cell function affecting the balance of peripheral immune responses against islet autoantigens, resulting in local inflammation in pancreatic islets.
Collapse
Affiliation(s)
- Tania Singh
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden.,Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Jesper K Colberg
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden
| | - Luis Sarmiento
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Patricia Chaves
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden
| | - Lisbeth Hansen
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Sara Bsharat
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden.,Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Luis R Cataldo
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden.,Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | | | - Malin Fex
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - David Bryder
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden
| | - Dan Holmberg
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Ewa Sitnicka
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden
| | - Corrado Cilio
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Rashmi B Prasad
- Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden
| | - Isabella Artner
- Stem Cell Center, Lund University, Klinikgatan 26, Lund, 22184, Sweden. .,Lund University Diabetes Center, Jan Waldenströms gata 35, Malmö, 21428, Sweden.
| |
Collapse
|