51
|
Impairment of a distinct cancer-associated fibroblast population limits tumour growth and metastasis. Nat Commun 2021; 12:3516. [PMID: 34112782 PMCID: PMC8192501 DOI: 10.1038/s41467-021-23583-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Profiling studies have revealed considerable phenotypic heterogeneity in cancer-associated fibroblasts (CAFs) present within the tumour microenvironment, however, functional characterisation of different CAF subsets is hampered by the lack of specific markers defining these populations. Here we show that genetic deletion of the Endo180 (MRC2) receptor, predominantly expressed by a population of matrix-remodelling CAFs, profoundly limits tumour growth and metastasis; effects that can be recapitulated in 3D co-culture assays. This impairment results from a CAF-intrinsic contractility defect and reduced CAF viability, which coupled with the lack of phenotype in the normal mouse, demonstrates that upregulated Endo180 expression by a specific, potentially targetable CAF subset is required to generate a supportive tumour microenvironment. Further, characterisation of a tumour subline selected via serial in vivo passage for its ability to overcome these stromal defects provides important insight into, how tumour cells adapt to a non-activated stroma in the early stages of metastatic colonisation. Endo180, a collagen binding receptor, is highly expressed in a subset of cancer-associated fibroblasts. The authors show, using knockout mice and 3D in vitro assays, that Endo180 depletion impairs tumour fibroblast contractility and viability resulting in reduced tumour growth and metastasis.
Collapse
|
52
|
Jacquemin G, Benavente-Diaz M, Djaber S, Bore A, Dangles-Marie V, Surdez D, Tajbakhsh S, Fre S, Lloyd-Lewis B. Longitudinal high-resolution imaging through a flexible intravital imaging window. SCIENCE ADVANCES 2021; 7:7/25/eabg7663. [PMID: 34134982 PMCID: PMC8208712 DOI: 10.1126/sciadv.abg7663] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 04/30/2021] [Indexed: 05/03/2023]
Abstract
Intravital microscopy (IVM) is a powerful technique that enables imaging of internal tissues at (sub)cellular resolutions in living animals. Here, we present a silicone-based imaging window consisting of a fully flexible, sutureless design that is ideally suited for long-term, longitudinal IVM of growing tissues and tumors. Crucially, we show that this window, without any customization, is suitable for numerous anatomical locations in mice using a rapid and standardized implantation procedure. This low-cost device represents a substantial technological and performance advance that facilitates intravital imaging in diverse contexts in higher organisms, opening previously unattainable avenues for in vivo imaging of soft and fragile tissues.
Collapse
Affiliation(s)
- Guillaume Jacquemin
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, F-75248 Paris Cedex 05, France.
| | - Maria Benavente-Diaz
- Stem Cells & Development Unit, Institut Pasteur, 25 rue du Dr. Roux, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
- Sorbonne Universités, Complexité du Vivant, F-75005, Paris, France
| | - Samir Djaber
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, F-75248 Paris Cedex 05, France
| | - Aurélien Bore
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, F-75248 Paris Cedex 05, France
- CRISPR'it, Platform for Genetic Screens, Institut Curie, PSL Research University, INSERM U934, CNRS UMR3215, F-75248 Paris Cedex 05, France
| | - Virginie Dangles-Marie
- Faculty of Pharmacy, Université Paris Descartes, Paris, France
- In vivo Experiment Platform, PSL Research University, 75005 Paris, France
| | - Didier Surdez
- INSERM U830, Équipe Labellisée LNCC, Diversity and Plasticity of Childhood Tumors Lab, PSL Research University, SIREDO Oncology Centre, Institut Curie Research Centre, Paris, France
| | - Shahragim Tajbakhsh
- Stem Cells & Development Unit, Institut Pasteur, 25 rue du Dr. Roux, 75015 Paris, France
- UMR CNRS 3738, Institut Pasteur, Paris, France
| | - Silvia Fre
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, F-75248 Paris Cedex 05, France.
| | - Bethan Lloyd-Lewis
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, F-75248 Paris Cedex 05, France.
- School of Cellular and Molecular Medicine, University of Bristol, Biomedical Sciences Building, Bristol, BS8 1TD, UK
| |
Collapse
|
53
|
Poscablo DM, Worthington AK, Smith-Berdan S, Forsberg EC. Megakaryocyte progenitor cell function is enhanced upon aging despite the functional decline of aged hematopoietic stem cells. Stem Cell Reports 2021; 16:1598-1613. [PMID: 34019813 PMCID: PMC8190594 DOI: 10.1016/j.stemcr.2021.04.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 04/23/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022] Open
Abstract
Age-related morbidity is associated with a decline in hematopoietic stem cell (HSC) function, but the mechanisms of HSC aging remain unclear. We performed heterochronic HSC transplants followed by quantitative analysis of cell reconstitution. Although young HSCs outperformed old HSCs in young recipients, young HSCs unexpectedly failed to outcompete the old HSCs of aged recipients. Interestingly, despite substantial enrichment of megakaryocyte progenitors (MkPs) in old mice in situ and reported platelet (Plt) priming with age, transplanted old HSCs were deficient in reconstitution of all lineages, including MkPs and Plts. We therefore performed functional analysis of young and old MkPs. Surprisingly, old MkPs displayed unmistakably greater regenerative capacity compared with young MkPs. Transcriptome analysis revealed putative molecular regulators of old MkP expansion. Collectively, these data demonstrated that aging affects HSCs and megakaryopoiesis in fundamentally different ways: whereas old HSCs functionally decline, MkPs gain expansion capacity upon aging. Reconstitution deficit by old HSCs was observed by chimerism and absolute cell numbers Young HSCs did not outcompete resident HSCs in aged recipient mice Old MkPs display remarkable capacity to engraft, expand, and reconstitute platelets Aging is associated with changes in MkP genome-wide expression signatures
Collapse
Affiliation(s)
- Donna M Poscablo
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, USA; Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Atesh K Worthington
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, USA; Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, USA; Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, USA.
| |
Collapse
|
54
|
Germline heterozygous mutations in Nxf1 perturb RNA metabolism and trigger thrombocytopenia and lymphopenia in mice. Blood Adv 2021; 4:1270-1283. [PMID: 32236527 DOI: 10.1182/bloodadvances.2019001323] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
In eukaryotic cells, messenger RNA (mRNA) molecules are exported from the nucleus to the cytoplasm, where they are translated. The highly conserved protein nuclear RNA export factor1 (Nxf1) is an important mediator of this process. Although studies in yeast and in human cell lines have shed light on the biochemical mechanisms of Nxf1 function, its contribution to mammalian physiology is less clear. Several groups have identified recurrent NXF1 mutations in chronic lymphocytic leukemia (CLL), placing it alongside several RNA-metabolism factors (including SF3B1, XPO, RPS15) whose dysregulation is thought to contribute to CLL pathogenesis. We report here an allelic series of germline point mutations in murine Nxf1. Mice heterozygous for these loss-of-function Nxf1 mutations exhibit thrombocytopenia and lymphopenia, together with milder hematological defects. This is primarily caused by cell-intrinsic defects in the survival of platelets and peripheral lymphocytes, which are sensitized to intrinsic apoptosis. In contrast, Nxf1 mutations have almost no effect on red blood cell homeostasis. Comparative transcriptome analysis of platelets, lymphocytes, and erythrocytes from Nxf1-mutant mice shows that, in response to impaired Nxf1 function, the cytoplasmic representation of transcripts encoding regulators of RNA metabolism is altered in a unique, lineage-specific way. Thus, blood cell lineages exhibit differential requirements for Nxf1-mediated global mRNA export.
Collapse
|
55
|
JAK2-V617F and interferon-α induce megakaryocyte-biased stem cells characterized by decreased long-term functionality. Blood 2021; 137:2139-2151. [PMID: 33667305 DOI: 10.1182/blood.2020005563] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
We studied a subset of hematopoietic stem cells (HSCs) that are defined by elevated expression of CD41 (CD41hi) and showed bias for differentiation toward megakaryocytes (Mks). Mouse models of myeloproliferative neoplasms (MPNs) expressing JAK2-V617F (VF) displayed increased frequencies and percentages of the CD41hi vs CD41lo HSCs compared with wild-type controls. An increase in CD41hi HSCs that correlated with JAK2-V617F mutant allele burden was also found in bone marrow from patients with MPN. CD41hi HSCs produced a higher number of Mk-colonies of HSCs in single-cell cultures in vitro, but showed reduced long-term reconstitution potential compared with CD41lo HSCs in competitive transplantations in vivo. RNA expression profiling showed an upregulated cell cycle, Myc, and oxidative phosphorylation gene signatures in CD41hi HSCs, whereas CD41lo HSCs showed higher gene expression of interferon and the JAK/STAT and TNFα/NFκB signaling pathways. Higher cell cycle activity and elevated levels of reactive oxygen species were confirmed in CD41hi HSCs by flow cytometry. Expression of Epcr, a marker for quiescent HSCs inversely correlated with expression of CD41 in mice, but did not show such reciprocal expression pattern in patients with MPN. Treatment with interferon-α further increased the frequency and percentage of CD41hi HSCs and reduced the number of JAK2-V617F+ HSCs in mice and patients with MPN. The shift toward the CD41hi subset of HSCs by interferon-α provides a possible mechanism of how interferon-α preferentially targets the JAK2 mutant clone.
Collapse
|
56
|
Chavez JS, Rabe JL, Loeffler D, Higa KC, Hernandez G, Mills TS, Ahmed N, Gessner RL, Ke Z, Idler BM, Niño KE, Kim H, Myers JR, Stevens BM, Davizon-Castillo P, Jordan CT, Nakajima H, Ashton J, Welner RS, Schroeder T, DeGregori J, Pietras EM. PU.1 enforces quiescence and limits hematopoietic stem cell expansion during inflammatory stress. J Exp Med 2021; 218:211996. [PMID: 33857288 PMCID: PMC8056754 DOI: 10.1084/jem.20201169] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 02/01/2021] [Accepted: 03/17/2021] [Indexed: 12/27/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are capable of entering the cell cycle to replenish the blood system in response to inflammatory cues; however, excessive proliferation in response to chronic inflammation can lead to either HSC attrition or expansion. The mechanism(s) that limit HSC proliferation and expansion triggered by inflammatory signals are poorly defined. Here, we show that long-term HSCs (HSCLT) rapidly repress protein synthesis and cell cycle genes following treatment with the proinflammatory cytokine interleukin (IL)-1. This gene program is associated with activation of the transcription factor PU.1 and direct PU.1 binding at repressed target genes. Notably, PU.1 is required to repress cell cycle and protein synthesis genes, and IL-1 exposure triggers aberrant protein synthesis and cell cycle activity in PU.1-deficient HSCs. These features are associated with expansion of phenotypic PU.1-deficient HSCs. Thus, we identify a PU.1-dependent mechanism triggered by innate immune stimulation that limits HSC proliferation and pool size. These findings provide insight into how HSCs maintain homeostasis during inflammatory stress.
Collapse
Affiliation(s)
- James S Chavez
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jennifer L Rabe
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Dirk Loeffler
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Kelly C Higa
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Giovanny Hernandez
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Taylor S Mills
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Nouraiz Ahmed
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Rachel L Gessner
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Zhonghe Ke
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Beau M Idler
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Katia E Niño
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Hyunmin Kim
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jason R Myers
- Genomics Research Center, University of Rochester, Rochester, NY
| | - Brett M Stevens
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | - Craig T Jordan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Hideaki Nakajima
- Department of Stem Cell and Immune Regulation, Yokohama City University School of Medicine, Yokohama, Japan
| | - John Ashton
- Genomics Research Center, University of Rochester, Rochester, NY
| | - Robert S Welner
- Division of Hematology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - James DeGregori
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO.,Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
57
|
Nakamura Y, Jörg DJ, Kon Y, Simons BD, Yoshida S. Transient suppression of transplanted spermatogonial stem cell differentiation restores fertility in mice. Cell Stem Cell 2021; 28:1443-1456.e7. [PMID: 33848470 PMCID: PMC8351876 DOI: 10.1016/j.stem.2021.03.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 12/16/2020] [Accepted: 03/19/2021] [Indexed: 01/11/2023]
Abstract
A remarkable feature of tissue stem cells is their ability to regenerate the structure and function of host tissue following transplantation. However, the dynamics of donor stem cells during regeneration remains largely unknown. Here we conducted quantitative clonal fate studies of transplanted mouse spermatogonial stem cells in host seminiferous tubules. We found that, after a large population of donor spermatogonia settle in host testes, through stochastic fate choice, only a small fraction persist and regenerate over the long term, and the rest are lost through differentiation and cell death. Further, based on these insights, we showed how repopulation efficiency can be increased to a level where the fertility of infertile hosts is restored by transiently suppressing differentiation using a chemical inhibitor of retinoic acid synthesis. These findings unlock a range of potential applications of spermatogonial transplantation, from fertility restoration in individuals with cancer to conservation of biological diversity.
Collapse
Affiliation(s)
- Yoshiaki Nakamura
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki 444-8787, Japan; Laboratory of Animal Breeding and Genetics, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-4-4 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8528, Japan; Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo 102-0083, Japan
| | - David J Jörg
- Wellcome Trust-Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Yayoi Kon
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki 444-8787, Japan
| | - Benjamin D Simons
- Wellcome Trust-Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Wilberforce Road, Cambridge CB3 0WA, UK; Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK.
| | - Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki 444-8787, Japan; Department of Basic Biology, School of Life Science, Graduate University for Advanced Studies (Sokendai), Okazaki 444-8787, Japan.
| |
Collapse
|
58
|
Di Pilato M, Palomino-Segura M, Mejías-Pérez E, Gómez CE, Rubio-Ponce A, D'Antuono R, Pizzagalli DU, Pérez P, Kfuri-Rubens R, Benguría A, Dopazo A, Ballesteros I, Sorzano COS, Hidalgo A, Esteban M, Gonzalez SF. Neutrophil subtypes shape HIV-specific CD8 T-cell responses after vaccinia virus infection. NPJ Vaccines 2021; 6:52. [PMID: 33846352 PMCID: PMC8041892 DOI: 10.1038/s41541-021-00314-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 03/09/2021] [Indexed: 12/20/2022] Open
Abstract
Neutrophils are innate immune cells involved in the elimination of pathogens and can also induce adaptive immune responses. Nα and Nβ neutrophils have been described with distinct in vitro capacity to generate antigen-specific CD8 T-cell responses. However, how these cell types exert their role in vivo and how manipulation of Nβ/Nα ratio influences vaccine-mediated immune responses are not known. In this study, we find that these neutrophil subtypes show distinct migratory and motility patterns and different ability to interact with CD8 T cells in the spleen following vaccinia virus (VACV) infection. Moreover, after analysis of adhesion, inflammatory, and migration markers, we observe that Nβ neutrophils overexpress the α4β1 integrin compared to Nα. Finally, by inhibiting α4β1 integrin, we increase the Nβ/Nα ratio and enhance CD8 T-cell responses to HIV VACV-delivered antigens. These findings provide significant advancements in the comprehension of neutrophil-based control of adaptive immune system and their relevance in vaccine design.
Collapse
Affiliation(s)
- Mauro Di Pilato
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland. .,Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain. .,Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA. .,Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Miguel Palomino-Segura
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Ernesto Mejías-Pérez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain.,Max von Pettenkofer-Institute, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Carmen E Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain
| | - Andrea Rubio-Ponce
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Rocco D'Antuono
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Crick Advanced Light Microscopy Science and Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Diego Ulisse Pizzagalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Institute of Computational Science, Università della Svizzera Italiana, Lugano, Switzerland
| | - Patricia Pérez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain
| | - Raphael Kfuri-Rubens
- Center of Integrated Protein Science Munich and Division of Clinical Pharmacology, Klinikum der Universität München, Munich, Germany
| | - Alberto Benguría
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Iván Ballesteros
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Carlos Oscar S Sorzano
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain
| | - Andrés Hidalgo
- Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología-CSIC, Madrid, Spain.
| | - Santiago F Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.
| |
Collapse
|
59
|
Monocyte-derived S1P in the lymph node regulates immune responses. Nature 2021; 592:290-295. [PMID: 33658712 PMCID: PMC8475585 DOI: 10.1038/s41586-021-03227-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/11/2021] [Indexed: 12/31/2022]
Abstract
The lipid chemoattractant sphingosine 1-phosphate (S1P) guides cells from the low-S1P environment of tissues into the high-S1P environment of circulatory fluids(1). Notably, S1P directs T cell exit from lymph nodes (LN), where T cells are initially activated, into lymph, from which T cells reach blood and ultimately inflamed tissues(1). T cells follow S1P gradients primarily using S1P receptor 1 (S1PR1)(1). While recent work has described how S1P gradients are established at steady-state, little is known about S1P distribution in disease, or about how changing S1P levels may affect immune responses. Here, we find that S1P concentrations increase in LN during an immune response. Hematopoietic cells, including inflammatory monocytes (iMo), are an important source of this S1P, an unexpected finding as endothelial cells provide lymph S1P(1). iMo require the early activation marker CD69 to supply this S1P, in part because CD69 expression is associated with reduced levels of S1pr5. CD69 acts as a “stand-your-ground” signal, keeping immune cells at a site of inflammation by regulating both S1P receptors and S1P gradients. Finally, increased S1P prolongs T cell residence time in LN, and exacerbates the severity of experimental autoimmune encephalomyelitis. This finding suggests the hypothesis that LN residence time regulates T cell differentiation, and points to novel uses of drugs targeting S1P signaling.
Collapse
|
60
|
Höpner SS, Raykova A, Radpour R, Amrein MA, Koller D, Baerlocher GM, Riether C, Ochsenbein AF. LIGHT/LTβR signaling regulates self-renewal and differentiation of hematopoietic and leukemia stem cells. Nat Commun 2021; 12:1065. [PMID: 33594067 PMCID: PMC7887212 DOI: 10.1038/s41467-021-21317-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/17/2021] [Indexed: 12/30/2022] Open
Abstract
The production of blood cells during steady-state and increased demand depends on the regulation of hematopoietic stem cell (HSC) self-renewal and differentiation. Similarly, the balance between self-renewal and differentiation of leukemia stem cells (LSCs) is crucial in the pathogenesis of leukemia. Here, we document that the TNF receptor superfamily member lymphotoxin-β receptor (LTβR) and its ligand LIGHT regulate quiescence and self-renewal of murine and human HSCs and LSCs. Cell-autonomous LIGHT/LTβR signaling on HSCs reduces cell cycling, promotes symmetric cell division and prevents primitive HSCs from exhaustion in serial re-transplantation experiments and genotoxic stress. LTβR deficiency reduces the numbers of LSCs and prolongs survival in a murine chronic myeloid leukemia (CML) model. Similarly, LIGHT/LTβR signaling in human G-CSF mobilized HSCs and human LSCs results in increased colony forming capacity in vitro. Thus, our results define LIGHT/LTβR signaling as an important pathway in the regulation of the self-renewal of HSCs and LSCs.
Collapse
MESH Headings
- Animals
- Antigens, CD34/metabolism
- Cell Cycle/drug effects
- Cell Cycle/genetics
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cell Self Renewal/drug effects
- Cell Self Renewal/genetics
- DNA Damage
- Fluorouracil/pharmacology
- Gene Expression Regulation, Leukemic/drug effects
- Hematopoietic Stem Cells/drug effects
- Hematopoietic Stem Cells/metabolism
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Lymphotoxin beta Receptor/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction/drug effects
- Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
- Mice
Collapse
Affiliation(s)
- S S Höpner
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ana Raykova
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - R Radpour
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - M A Amrein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - D Koller
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - G M Baerlocher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - C Riether
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - A F Ochsenbein
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
61
|
Soares-da-Silva F, Freyer L, Elsaid R, Burlen-Defranoux O, Iturri L, Sismeiro O, Pinto-do-Ó P, Gomez-Perdiguero E, Cumano A. Yolk sac, but not hematopoietic stem cell-derived progenitors, sustain erythropoiesis throughout murine embryonic life. J Exp Med 2021; 218:211777. [PMID: 33566111 PMCID: PMC7879581 DOI: 10.1084/jem.20201729] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/13/2020] [Accepted: 01/07/2021] [Indexed: 01/03/2023] Open
Abstract
In the embryo, the first hematopoietic cells derive from the yolk sac and are thought to be rapidly replaced by the progeny of hematopoietic stem cells. We used three lineage-tracing mouse models to show that, contrary to what was previously assumed, hematopoietic stem cells do not contribute significantly to erythrocyte production up until birth. Lineage tracing of yolk sac erythromyeloid progenitors, which generate tissue resident macrophages, identified highly proliferative erythroid progenitors that rapidly differentiate after intra-embryonic injection, persisting as the major contributors to the embryonic erythroid compartment. We show that erythrocyte progenitors of yolk sac origin require 10-fold lower concentrations of erythropoietin than their hematopoietic stem cell–derived counterparts for efficient erythrocyte production. We propose that, in a low erythropoietin environment in the fetal liver, yolk sac–derived erythrocyte progenitors efficiently outcompete hematopoietic stem cell progeny, which fails to generate megakaryocyte and erythrocyte progenitors.
Collapse
Affiliation(s)
- Francisca Soares-da-Silva
- Lymphocytes and Immunity Unit, Immunology Department, Institut National de la Santé et de la Recherche Médicale U1223, Institut Pasteur, Paris, France.,Instituto de Investigação e Inovação em Saúde and Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Graduate Program in Areas of Basic and Applied Biology, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Laina Freyer
- Macrophages and Endothelial Cells, Department of Developmental and Stem Cell Biology, UMR3738 Centre national de la recherche scientifique, Institut Pasteur, Paris, France
| | - Ramy Elsaid
- Lymphocytes and Immunity Unit, Immunology Department, Institut National de la Santé et de la Recherche Médicale U1223, Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Odile Burlen-Defranoux
- Lymphocytes and Immunity Unit, Immunology Department, Institut National de la Santé et de la Recherche Médicale U1223, Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Lorea Iturri
- Macrophages and Endothelial Cells, Department of Developmental and Stem Cell Biology, UMR3738 Centre national de la recherche scientifique, Institut Pasteur, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Odile Sismeiro
- Institut Pasteur, Transcriptome and EpiGenome, Biomics Center for Innovation and Technological Research, Paris, France
| | - Perpétua Pinto-do-Ó
- Instituto de Investigação e Inovação em Saúde and Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Elisa Gomez-Perdiguero
- Macrophages and Endothelial Cells, Department of Developmental and Stem Cell Biology, UMR3738 Centre national de la recherche scientifique, Institut Pasteur, Paris, France
| | - Ana Cumano
- Lymphocytes and Immunity Unit, Immunology Department, Institut National de la Santé et de la Recherche Médicale U1223, Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| |
Collapse
|
62
|
Stetka J, Skoda RC. Mouse models of myeloproliferative neoplasms for pre-clinical testing of novel therapeutic agents. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2021; 165:26-33. [PMID: 33542546 DOI: 10.5507/bp.2021.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 11/23/2022] Open
Abstract
Myeloproliferative neoplasms (MPN), are clonal hematopoietic stem cell (HSC) disorders driven by gain-of-function mutations in JAK2 (JAK2-V617F), CALR or MPL genes. MPN treatment options currently mainly consist of cytoreductive therapy with hydroxyurea and JAK2 inhibitors such as ruxolitinib and fedratinib. Pegylated interferon-alpha can induce complete molecular remission (CMR) in some MPN patients when applied at early stages of disease. The ultimate goal of modern MPN treatment is to develop novel therapies that specifically target mutant HSCs in MPN and consistently induce CMR. Basic research has identified a growing number of candidate drugs with promising effects in vitro. A first step on the way to developing these compounds into drugs approved for treatment of MPN patients often consists of examining the effects in vivo using pre-clinical mouse models of MPN. Here we review the current state of MPN mouse models and the experimental setup for their optimal use in drug testing. In addition to novel compounds, combinatorial therapeutic approaches are often considered for the treatment of MPN. Optimized and validated mouse models can provide an efficient way to rapidly assess and select the most promising combinations and thereby contribute to accelerating the development of novel therapies of MPN.
Collapse
Affiliation(s)
- Jan Stetka
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland.,Department of Biology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | - Radek C Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
63
|
Yu X, Persillet M, Zhang L, Zhang Y, Xiuping S, Li X, Ran G, Breger LS, Dovero S, Porras G, Dehay B, Bezard E, Qin C. Evaluation of blood flow as a route for propagation in experimental synucleinopathy. Neurobiol Dis 2021; 150:105255. [PMID: 33421564 DOI: 10.1016/j.nbd.2021.105255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/02/2021] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
In Parkinson's disease, synucleinopathy is hypothesized to spread from the enteric nervous system, via the vagus nerve, to the central nervous system. Recent evidences collected in non-human primates challenge however the hypothesis of a transmission of α-synuclein (α-syn) pathology through the vagus nerve. Would the hypothesis whereby the bloodstream acts as a route for long-distance transmission of pathological α-syn hold true, an inter-individual transmission of synucleinopathy could occur via blood contact. Here, we used a parabiosis approach to join the circulatory systems of wild type and GFP transgenic C57BL/6 J mice, for which one of the partners parabiont received a stereotaxic intranigral injection of patient-derived α-syn aggregates. While the Lewy Body-receiving mice exhibited a loss of dopamine neurons and an increase in nigral S129 phosphorylated α-syn immunoreactivity, their parabiotic bloodstream-sharing partners did not show any trend for a lesion or change in S129 phosphorylated-α-syn levels. Altogether, our study suggests that, in the patient-derived α-synuclein aggregates-injected mouse model and within the selected time frame, the disease is not "transmitted" through the bloodstream.
Collapse
Affiliation(s)
- Xuan Yu
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | | | - Ling Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Yu Zhang
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Sun Xiuping
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Xianglei Li
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | - Gao Ran
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China
| | | | - Sandra Dovero
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Gregory Porras
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Benjamin Dehay
- Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France
| | - Erwan Bezard
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China; Univ. Bordeaux, CNRS, IMN, UMR 5293, F-33000 Bordeaux, France.
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Peking Union Medical College (PUMC) & Institute of Laboratory Animal Science, Chinese Academy of Medical Science (CAMS), Beijing, China.
| |
Collapse
|
64
|
The Clot Thickens: Recent Clues on Hematopoietic Stem Cell Contribution to Age-Related Platelet Biology Open New Questions. ADVANCES IN GERIATRIC MEDICINE AND RESEARCH 2021; 3. [PMID: 35037001 PMCID: PMC8759758 DOI: 10.20900/agmr20210019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Platelets provide life-saving functions by halting external and internal bleeding. There is also a dark side to platelet biology, however. Recent reports provide evidence for increased platelet reactivity during aging of mice and humans, making platelets main suspects in the most prevalent aging-related human pathologies, including cardiovascular diseases, stroke, and cancer. What drives this platelet hyperreactivity during aging? Here, we discuss how hematopoietic stem cell differentiation pathways into the platelet lineage offer avenues to understand the fundamental differences between young and old platelets. Recent advances begin to unravel how the cellular and molecular regulation of the parent hematopoietic stem and progenitor cells likely imbue aging characteristics on the resulting Plt progeny. The resulting mechanistic insights into intrinsic platelet reactivity will provide strategies for selectively targeting age-related pathways. This brief viewpoint focuses on current concepts on aging hematopoiesis and the implications for platelet hyperactivity during aging.
Collapse
|
65
|
Nečas E, Faltusová K, Chen CL. Latent Defect in Haematopoiesis of UBC-GFP Mice Sheds Light on the Lymphoid Developmental Potential of Haematopoietic Stem Cells. Folia Biol (Praha) 2021; 67:135. [PMID: 35151237 DOI: 10.14712/fb2021067040135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Affiliation(s)
- E Nečas
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Czech Republic
| | - K Faltusová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Czech Republic
| | - C-L Chen
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Czech Republic
| |
Collapse
|
66
|
Stolp B, Thelen F, Ficht X, Altenburger LM, Ruef N, Inavalli VVGK, Germann P, Page N, Moalli F, Raimondi A, Keyser KA, Seyed Jafari SM, Barone F, Dettmer MS, Merkler D, Iannacone M, Sharpe J, Schlapbach C, Fackler OT, Nägerl UV, Stein JV. Salivary gland macrophages and tissue-resident CD8 + T cells cooperate for homeostatic organ surveillance. Sci Immunol 2020; 5:5/46/eaaz4371. [PMID: 32245888 DOI: 10.1126/sciimmunol.aaz4371] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/10/2020] [Indexed: 01/26/2023]
Abstract
It is well established that tissue macrophages and tissue-resident memory CD8+ T cells (TRM) play important roles for pathogen sensing and rapid protection of barrier tissues. In contrast, the mechanisms by which these two cell types cooperate for homeostatic organ surveillance after clearance of infections is poorly understood. Here, we used intravital imaging to show that TRM dynamically followed tissue macrophage topology in noninflamed murine submandibular salivary glands (SMGs). Depletion of tissue macrophages interfered with SMG TRM motility and caused a reduction of interepithelial T cell crossing. In the absence of macrophages, SMG TRM failed to cluster in response to local inflammatory chemokines. A detailed analysis of the SMG microarchitecture uncovered discontinuous attachment of tissue macrophages to neighboring epithelial cells, with occasional macrophage protrusions bridging adjacent acini and ducts. When dissecting the molecular mechanisms that drive homeostatic SMG TRM motility, we found that these cells exhibit a wide range of migration modes: In addition to chemokine- and adhesion receptor-driven motility, resting SMG TRM displayed a remarkable capacity for autonomous motility in the absence of chemoattractants and adhesive ligands. Autonomous SMG TRM motility was mediated by friction and insertion of protrusions into gaps offered by the surrounding microenvironment. In sum, SMG TRM display a unique continuum of migration modes, which are supported in vivo by tissue macrophages to allow homeostatic patrolling of the complex SMG architecture.
Collapse
Affiliation(s)
- Bettina Stolp
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland.,Department for Infectious Diseases, Integrative Virology, Center for Integrative Infectious Disease Research, University Hospital Heidelberg, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany
| | - Flavian Thelen
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Xenia Ficht
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Lukas M Altenburger
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - V V G Krishna Inavalli
- University of Bordeaux, 33700 Bordeaux, France.,Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33077 Bordeaux, France
| | - Philipp Germann
- EMBL Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain.,Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain
| | - Nicolas Page
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospitals of Geneva, 1211 Geneva, Switzerland
| | | | | | - Kirsten A Keyser
- Institute for Virology, OE5230, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - S Morteza Seyed Jafari
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Francesca Barone
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | | | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, University and University Hospitals of Geneva, 1211 Geneva, Switzerland
| | | | - James Sharpe
- EMBL Barcelona, Dr. Aiguader 88, 08003 Barcelona, Spain.,Universitat Pompeu Fabra (UPF), 08002 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Pg. Lluis Companys 23, 08010 Barcelona, Spain
| | - Christoph Schlapbach
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Oliver T Fackler
- Department for Infectious Diseases, Integrative Virology, Center for Integrative Infectious Disease Research, University Hospital Heidelberg, Im Neuenheimer Feld 344, 69120 Heidelberg, Germany
| | - U Valentin Nägerl
- University of Bordeaux, 33700 Bordeaux, France.,Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, 33077 Bordeaux, France
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland.
| |
Collapse
|
67
|
Koeniger T, Bell L, Mifka A, Enders M, Hautmann V, Mekala SR, Kirchner P, Ekici AB, Schulz C, Wörsdörfer P, Mencl S, Kleinschnitz C, Ergün S, Kuerten S. Bone marrow-derived myeloid progenitors in the leptomeninges of adult mice. STEM CELLS (DAYTON, OHIO) 2020; 39:227-239. [PMID: 33270951 DOI: 10.1002/stem.3311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/29/2020] [Accepted: 11/17/2020] [Indexed: 11/11/2022]
Abstract
Although the bone marrow contains most hematopoietic activity during adulthood, hematopoietic stem and progenitor cells can be recovered from various extramedullary sites. Cells with hematopoietic progenitor properties have even been reported in the adult brain under steady-state conditions, but their nature and localization remain insufficiently defined. Here, we describe a heterogeneous population of myeloid progenitors in the leptomeninges of adult C57BL/6 mice. This cell pool included common myeloid, granulocyte/macrophage, and megakaryocyte/erythrocyte progenitors. Accordingly, it gave rise to all major myelo-erythroid lineages in clonogenic culture assays. Brain-associated progenitors persisted after tissue perfusion and were partially inaccessible to intravenous antibodies, suggesting their localization behind continuous blood vessel endothelium such as the blood-arachnoid barrier. Flt3Cre lineage tracing and bone marrow transplantation showed that the precursors were derived from adult hematopoietic stem cells and were most likely continuously replaced via cell trafficking. Importantly, their occurrence was tied to the immunologic state of the central nervous system (CNS) and was diminished in the context of neuroinflammation and ischemic stroke. Our findings confirm the presence of myeloid progenitors at the meningeal border of the brain and lay the foundation to unravel their possible functions in CNS surveillance and local immune cell production.
Collapse
Affiliation(s)
- Tobias Koeniger
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Luisa Bell
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Anika Mifka
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Enders
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Valentin Hautmann
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Subba Rao Mekala
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Ludwig Maximilian University of Munich, Munich, Germany
| | - Philipp Wörsdörfer
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Stine Mencl
- University Hospital Essen, Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Christoph Kleinschnitz
- University Hospital Essen, Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Julius Maximilian University of Würzburg, Würzburg, Germany.,Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.,Anatomisches Institut, Neuroanatomie, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
68
|
Brinkmann K, Waring P, Glaser SP, Wimmer V, Cottle DL, Tham MS, Nhu D, Whitehead L, Delbridge AR, Lessene G, Smyth IM, Herold MJ, Kelly GL, Grabow S, Strasser A. BCL-XL exerts a protective role against anemia caused by radiation-induced kidney damage. EMBO J 2020; 39:e105561. [PMID: 33236795 DOI: 10.15252/embj.2020105561] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/06/2020] [Accepted: 10/15/2020] [Indexed: 12/29/2022] Open
Abstract
Studies of gene-targeted mice identified the roles of the different pro-survival BCL-2 proteins during embryogenesis. However, little is known about the role(s) of these proteins in adults in response to cytotoxic stresses, such as treatment with anti-cancer agents. We investigated the role of BCL-XL in adult mice using a strategy where prior bone marrow transplantation allowed for loss of BCL-XL exclusively in non-hematopoietic tissues to prevent anemia caused by BCL-XL deficiency in erythroid cells. Unexpectedly, the combination of total body γ-irradiation (TBI) and genetic loss of Bcl-x caused secondary anemia resulting from chronic renal failure due to apoptosis of renal tubular epithelium with secondary obstructive nephropathy. These findings identify a critical protective role of BCL-XL in the adult kidney and inform on the use of BCL-XL inhibitors in combination with DNA damage-inducing drugs for cancer therapy. Encouragingly, the combination of DNA damage-inducing anti-cancer therapy plus a BCL-XL inhibitor could be tolerated in mice, at least when applied sequentially.
Collapse
Affiliation(s)
- Kerstin Brinkmann
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Paul Waring
- Department of Surgery, University of Melbourne, Melbourne, Vic., Australia
| | - Stefan P Glaser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Verena Wimmer
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Denny L Cottle
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Vic., Australia
| | - Ming Shen Tham
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Vic., Australia
| | - Duong Nhu
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Alex Rd Delbridge
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Guillaume Lessene
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Development and Stem Cell Program Monash Biomedicine Discovery Institute (BDI), Monash University, Melbourne, Vic., Australia.,Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Vic., Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Stephanie Grabow
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Vic., Australia.,Department of Medical Biology, University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
69
|
Belanger MC, Zhuang M, Ball AG, Richey KH, DeRosa CA, Fraser CL, Pompano RR. Labelling primary immune cells using bright blue fluorescent nanoparticles. Biomater Sci 2020; 8:1897-1909. [PMID: 32026891 DOI: 10.1039/c9bm01572h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tracking cell movements is an important aspect of many biological studies. Reagents for cell tracking must not alter the biological state of the cell and must be bright enough to be visualized above background autofluorescence, a particular concern when imaging in tissue. Currently there are few reagents compatible with standard UV excitation filter sets (e.g. DAPI) that fulfill those requirements, despite the development of many dyes optimized for violet excitation (405 nm). A family of boron-based fluorescent dyes, difluoroboron β-diketonates, has previously served as bio-imaging reagents with UV excitation, offering high quantum yields and wide excitation peaks. In this study, we investigated the use of one such dye as a potential cell tracking reagent. A library of difluoroboron dibenzoylmethane (BF2dbm) conjugates were synthesized with biocompatible polymers including: poly(l-lactic acid) (PLLA), poly(ε-caprolactone) (PCL), and block copolymers with poly(ethylene glycol) (PEG). Dye-polymer conjugates were fabricated into nanoparticles, which were stable for a week at 37 °C in water and cell culture media, but quickly aggregated in saline. Nanoparticles were used to label primary splenocytes; phagocytic cell types were more effectively labelled. Labelling with nanoparticles did not affect cellular viability, nor basic immune responses. Labelled cells were more easily distinguished when imaged on a live tissue background than those labelled with a commercially available UV-excitable cytoplasmic labelling reagent. The high efficiency in terms of both fluorescence and cellular labelling may allow these nanoparticles to act as a short-term cell labelling strategy while wide excitation peaks offer utility across imaging and analysis platforms.
Collapse
Affiliation(s)
- Maura C Belanger
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA. and Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Meng Zhuang
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA.
| | - Alexander G Ball
- Department of Microbiology Cancer Biology and Immunology, University of Virginia, Charlottesville, Virginia 22903, USA and Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22903, USA
| | - Kristen H Richey
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA.
| | - Christopher A DeRosa
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA.
| | - Cassandra L Fraser
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA.
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA. and Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia 22903, USA
| |
Collapse
|
70
|
Benlabiod C, Cacemiro MDC, Nédélec A, Edmond V, Muller D, Rameau P, Touchard L, Gonin P, Constantinescu SN, Raslova H, Villeval JL, Vainchenker W, Plo I, Marty C. Calreticulin del52 and ins5 knock-in mice recapitulate different myeloproliferative phenotypes observed in patients with MPN. Nat Commun 2020; 11:4886. [PMID: 32985500 PMCID: PMC7522233 DOI: 10.1038/s41467-020-18691-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 09/04/2020] [Indexed: 12/20/2022] Open
Abstract
Somatic mutations in the calreticulin (CALR) gene are associated with approximately 30% of essential thrombocythemia (ET) and primary myelofibrosis (PMF). CALR mutations, including the two most frequent 52 bp deletion (del52) and 5 bp insertion (ins5), induce a frameshift to the same alternative reading frame generating new C-terminal tails. In patients, del52 and ins5 induce two phenotypically distinct myeloproliferative neoplasms (MPNs). They are equally found in ET, but del52 is more frequent in PMF. We generated heterozygous and homozygous conditional inducible knock-in (KI) mice expressing a chimeric murine CALR del52 or ins5 with the human mutated C-terminal tail to investigate their pathogenic effects on hematopoiesis. Del52 induces greater phenotypic changes than ins5 including thrombocytosis, leukocytosis, splenomegaly, bone marrow hypocellularity, megakaryocytic lineage amplification, expansion and competitive advantage of the hematopoietic stem cell compartment. Homozygosity amplifies these features, suggesting a distinct contribution of homozygous clones to human MPNs. Moreover, homozygous del52 KI mice display features of a penetrant myelofibrosis-like disorder with extramedullary hematopoiesis linked to splenomegaly, megakaryocyte hyperplasia and the presence of reticulin fibers. Overall, modeling del52 and ins5 mutations in mice successfully recapitulates the differences in phenotypes observed in patients. Calreticulin del52 and ins5 mutations induce two phenotypically distinct myeloproliferative neoplasms in patients. Here the authors show that modeling these mutations in knock-in mice recapitulate the two diseases and highlight how they impact the different hematopoietic compartments.
Collapse
Affiliation(s)
- Camélia Benlabiod
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Maira da Costa Cacemiro
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France.,Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo-USP, Ribeirão Preto, São Paulo, Brazil
| | - Audrey Nédélec
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Brussels, Belgium
| | - Valérie Edmond
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Delphine Muller
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Philippe Rameau
- Integrated Biology Core Facility, Gustave Roussy, Villejuif, France
| | - Laure Touchard
- Preclinical Research Plateform, Unité Mixte de Service AMMICA 3655/US 23, Gustave Roussy, Villejuif, France
| | - Patrick Gonin
- Preclinical Research Plateform, Unité Mixte de Service AMMICA 3655/US 23, Gustave Roussy, Villejuif, France
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research, Brussels, Belgium.,de Duve Institute, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Université catholique de Louvain, Brussels, Belgium
| | - Hana Raslova
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Jean-Luc Villeval
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - William Vainchenker
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France.,Gustave Roussy, UMR 1287, Villejuif, France
| | - Caroline Marty
- INSERM, UMR 1287, Gustave Roussy, Villejuif, France. .,Université Paris-Saclay, UMR 1287, Gustave Roussy, Villejuif, France. .,Gustave Roussy, UMR 1287, Villejuif, France.
| |
Collapse
|
71
|
Crowley L, Cambuli F, Aparicio L, Shibata M, Robinson BD, Xuan S, Li W, Hibshoosh H, Loda M, Rabadan R, Shen MM. A single-cell atlas of the mouse and human prostate reveals heterogeneity and conservation of epithelial progenitors. eLife 2020; 9:e59465. [PMID: 32915138 PMCID: PMC7529463 DOI: 10.7554/elife.59465] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 09/10/2020] [Indexed: 01/06/2023] Open
Abstract
Understanding the cellular constituents of the prostate is essential for identifying the cell of origin for prostate adenocarcinoma. Here, we describe a comprehensive single-cell atlas of the adult mouse prostate epithelium, which displays extensive heterogeneity. We observe distal lobe-specific luminal epithelial populations (LumA, LumD, LumL, and LumV), a proximally enriched luminal population (LumP) that is not lobe-specific, and a periurethral population (PrU) that shares both basal and luminal features. Functional analyses suggest that LumP and PrU cells have multipotent progenitor activity in organoid formation and tissue reconstitution assays. Furthermore, we show that mouse distal and proximal luminal cells are most similar to human acinar and ductal populations, that a PrU-like population is conserved between species, and that the mouse lateral prostate is most similar to the human peripheral zone. Our findings elucidate new prostate epithelial progenitors, and help resolve long-standing questions about anatomical relationships between the mouse and human prostate.
Collapse
Affiliation(s)
- Laura Crowley
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Francesco Cambuli
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Luis Aparicio
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Biomedical Informatics, Columbia University Irving Medical CenterNew YorkUnited States
| | - Maho Shibata
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell UniversityNew YorkUnited States
| | - Shouhong Xuan
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Weiping Li
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| | - Hanina Hibshoosh
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Pathology and Cell Biology, Columbia University Irving Medical CenterNew YorkUnited States
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell UniversityNew YorkUnited States
| | - Raul Rabadan
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Biomedical Informatics, Columbia University Irving Medical CenterNew YorkUnited States
| | - Michael M Shen
- Department of Medicine, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Genetics and Development, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Urology, Columbia University Irving Medical CenterNew YorkUnited States
- Department of Systems Biology, Columbia University Irving Medical CenterNew YorkUnited States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical CenterNew YorkUnited States
| |
Collapse
|
72
|
Wehn AK, Farkas DR, Sedlock CE, Subedi D, Chapman DL. Functionally distinct roles for T and Tbx6 during mouse development. Biol Open 2020; 9:9/8/bio054692. [PMID: 32855167 PMCID: PMC7473639 DOI: 10.1242/bio.054692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The mouse T-box transcription factors T and Tbx6 are co-expressed in the primitive streak and have unique domains of expression; T is expressed in the notochord, while Tbx6 is expressed in the presomitic mesoderm. T-box factors are related through a shared DNA binding domain, the T-domain, and can therefore bind to similar DNA sequences at least in vitro. We investigated the functional similarities and differences of T and Tbx6 DNA binding and transcriptional activity in vitro and their interaction genetically in vivo. We show that at one target, Dll1, the T-domains of T and Tbx6 have different affinities for the binding sites present in the mesoderm enhancer. We further show using in vitro assays that T and Tbx6 differentially affect transcription with Tbx6 activating expression tenfold higher than T, that T and Tbx6 can compete at target gene enhancers, and that this competition requires a functional DNA binding domain. Next, we addressed whether T and Tbx6 can compete in vivo. First, we generated embryos that express Tbx6 at greater than wild-type levels embryos and show that these embryos have short tails, resembling the T heterozygous phenotype. Next, using the dominant-negative TWis allele, we show that Tbx6+/− TWis/+ embryos share similarities with embryos homozygous for the Tbx6 hypomorphic allele rib-vertebrae, specifically fusions of several ribs and malformation of some vertebrae. Finally, we tested whether Tbx6 can functionally replace T using a knockin approach, which resulted in severe T null-like phenotypes in chimeric embryos generated with ES cells heterozygous for a Tbx6 knockin at the T locus. Altogether, our results of differences in affinity for DNA binding sites and transcriptional activity for T and Tbx6 provide a potential mechanism for the failure of Tbx6 to functionally replace T and possible competition phenotypes in vivo. Summary: Mouse Tbx6 fails to compensate for heterozygous loss of T; instead ectopic Tbx6 in the T expression-domain in knockin embryos generates T null-like phenotypes suggestive of competition.
Collapse
Affiliation(s)
- Amy K Wehn
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Deborah R Farkas
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Carly E Sedlock
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Dibya Subedi
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Deborah L Chapman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| |
Collapse
|
73
|
Kobayashi H, Morikawa T, Okinaga A, Hamano F, Hashidate-Yoshida T, Watanuki S, Hishikawa D, Shindou H, Arai F, Kabe Y, Suematsu M, Shimizu T, Takubo K. Environmental Optimization Enables Maintenance of Quiescent Hematopoietic Stem Cells Ex Vivo. Cell Rep 2020; 28:145-158.e9. [PMID: 31269436 DOI: 10.1016/j.celrep.2019.06.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/26/2018] [Accepted: 06/03/2019] [Indexed: 12/25/2022] Open
Abstract
Hematopoietic stem cells (HSCs) maintain lifelong hematopoiesis by remaining quiescent in the bone marrow niche. Recapitulation of a quiescent state in culture has not been achieved, as cells rapidly proliferate and differentiate in vitro. After exhaustive analysis of different environmental factor combinations and concentrations as a way to mimic physiological conditions, we were able to maintain engraftable quiescent HSCs for 1 month in culture under very low cytokine concentrations, hypoxia, and very high fatty acid levels. Exogenous fatty acids were required likely due to suppression of intrinsic fatty acid synthesis by hypoxia and low cytokine conditions. By contrast, high cytokine concentrations or normoxia induced HSC proliferation and differentiation. Our culture system provides a means to evaluate properties of steady-state HSCs and test effects of defined factors in vitro under near-physiological conditions.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Takayuki Morikawa
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Ayumi Okinaga
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Fumie Hamano
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Tomomi Hashidate-Yoshida
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Shintaro Watanuki
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan; Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Daisuke Hishikawa
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan; Department of Lipid Science, Graduate School of Medicine, The University of Tokyo, 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Fumio Arai
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yasuaki Kabe
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takao Shimizu
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan; Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Keiyo Takubo
- Department of Stem Cell Biology, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, Tokyo 162-8655, Japan.
| |
Collapse
|
74
|
Lee J, Chong T, Newmark PA. The esophageal gland mediates host immune evasion by the human parasite Schistosoma mansoni. Proc Natl Acad Sci U S A 2020; 117:19299-19309. [PMID: 32737161 PMCID: PMC7431036 DOI: 10.1073/pnas.2006553117] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Schistosomes are parasitic flatworms that cause schistosomiasis, a neglected tropical disease affecting over 200 million people. Schistosomes develop multiple body plans while navigating their complex life cycle, which involves two different hosts: a mammalian definitive host and a molluscan intermediate host. Their survival and propagation depend upon proliferation and differentiation of stem cells necessary for parasite homeostasis and reproduction. Infective larvae released from snails carry a handful of stem cells that serve as the likely source of new tissues as the parasite adapts to life inside the mammalian host; however, the role of these stem cells during this critical life cycle stage remains unclear. Here, we characterize stem cell fates during early intramammalian development. Surprisingly, we find that the esophageal gland, an accessory organ of the digestive tract, develops before the rest of the digestive system is formed and blood feeding is initiated, suggesting a role in processes beyond nutrient uptake. To explore such a role, we examine schistosomes that lack the esophageal gland due to knockdown of a forkhead-box transcription factor, Sm-foxA, which blocks development and maintenance of the esophageal gland, without affecting the development of other somatic tissues. Intriguingly, schistosomes lacking the esophageal gland die after transplantation into naive mice, but survive in immunodeficient mice lacking B cells. We show that parasites lacking the esophageal gland are unable to lyse ingested immune cells within the esophagus before passing them into the gut. These results unveil an immune-evasion mechanism mediated by the esophageal gland, which is essential for schistosome survival and pathogenesis.
Collapse
Affiliation(s)
- Jayhun Lee
- Regenerative Biology, Morgridge Institute for Research, Madison, WI 53715
| | - Tracy Chong
- Regenerative Biology, Morgridge Institute for Research, Madison, WI 53715
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53715
| | - Phillip A Newmark
- Regenerative Biology, Morgridge Institute for Research, Madison, WI 53715;
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53715
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI 53715
| |
Collapse
|
75
|
Lau D, Garçon F, Chandra A, Lechermann LM, Aloj L, Chilvers ER, Corrie PG, Okkenhaug K, Gallagher FA. Intravital Imaging of Adoptive T-Cell Morphology, Mobility and Trafficking Following Immune Checkpoint Inhibition in a Mouse Melanoma Model. Front Immunol 2020; 11:1514. [PMID: 32793206 PMCID: PMC7387409 DOI: 10.3389/fimmu.2020.01514] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Efficient T-cell targeting, infiltration and activation within tumors is crucial for successful adoptive T-cell therapy. Intravital microscopy is a powerful tool for the visualization of T-cell behavior within tumors, as well as spatial and temporal heterogeneity in response to immunotherapy. Here we describe an experimental approach for intravital imaging of adoptive T-cell morphology, mobility and trafficking in a skin-flap tumor model, following immune modulation with immune checkpoint inhibitors (ICIs) targeting PD-L1 and CTLA-4. A syngeneic model of ovalbumin and mCherry-expressing amelanotic mouse melanoma was used in conjunction with adoptively transferred OT-1+ cytotoxic T-cells expressing GFP to image antigen-specific live T-cell behavior within the tumor microenvironment. Dynamic image analysis of T-cell motility showed distinct CD8+ T-cell migration patterns and morpho-dynamics within different tumor compartments in response to ICIs: this approach was used to cluster T-cell behavior into four groups based on velocity and meandering index. The results showed that most T-cells within the tumor periphery demonstrated Lévy-like trajectories, consistent with tumor cell searching strategies. T-cells adjacent to tumor cells had reduced velocity and appeared to probe the local environment, consistent with cell-cell interactions. An increased number of T-cells were detected following treatment, traveling at lower mean velocities than controls, and demonstrating reduced displacement consistent with target engagement. Histogram-based analysis of immunofluorescent images from harvested tumors showed that in the ICI-treated mice there was a higher density of CD31+ vessels compared to untreated controls and a greater infiltration of T-cells towards the tumor core, consistent with increased cellular trafficking post-treatment.
Collapse
Affiliation(s)
- Doreen Lau
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| | - Fabien Garçon
- Laboratory of Lymphocyte Signaling and Development, The Babraham Institute, Cambridge, United Kingdom
| | - Anita Chandra
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | - Luigi Aloj
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
- Department of Nuclear Medicine, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Edwin R. Chilvers
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Pippa G. Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Ferdia A. Gallagher
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Department of Radiology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
76
|
Bellomo A, Mondor I, Spinelli L, Lagueyrie M, Stewart BJ, Brouilly N, Malissen B, Clatworthy MR, Bajénoff M. Reticular Fibroblasts Expressing the Transcription Factor WT1 Define a Stromal Niche that Maintains and Replenishes Splenic Red Pulp Macrophages. Immunity 2020; 53:127-142.e7. [PMID: 32562599 DOI: 10.1016/j.immuni.2020.06.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/20/2020] [Accepted: 06/04/2020] [Indexed: 12/21/2022]
Abstract
Located within red pulp cords, splenic red pulp macrophages (RPMs) are constantly exposed to the blood flow, clearing senescent red blood cells (RBCs) and recycling iron from hemoglobin. Here, we studied the mechanisms underlying RPM homeostasis, focusing on the involvement of stromal cells as these cells perform anchoring and nurturing macrophage niche functions in lymph nodes and liver. Microscopy revealed that RPMs are embedded in a reticular meshwork of red pulp fibroblasts characterized by the expression of the transcription factor Wilms' Tumor 1 (WT1) and colony stimulating factor 1 (CSF1). Conditional deletion of Csf1 in WT1+ red pulp fibroblasts, but not white pulp fibroblasts, drastically altered the RPM network without altering circulating CSF1 levels. Upon RPM depletion, red pulp fibroblasts transiently produced the monocyte chemoattractants CCL2 and CCL7, thereby contributing to the replenishment of the RPM network. Thus, red pulp fibroblasts anchor and nurture RPM, a function likely conserved in humans.
Collapse
Affiliation(s)
- Alicia Bellomo
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | | | | | | | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nicolas Brouilly
- Aix-Marseille Université, Centre National de la Recherche Scientifique, Institut de Biologie du Développement de Marseille, Marseille, France
| | | | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK; Cambridge University Hospitals NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge, UK; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Marc Bajénoff
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.
| |
Collapse
|
77
|
Wosczyna MN, Konishi CT, Perez Carbajal EE, Wang TT, Walsh RA, Gan Q, Wagner MW, Rando TA. Mesenchymal Stromal Cells Are Required for Regeneration and Homeostatic Maintenance of Skeletal Muscle. Cell Rep 2020; 27:2029-2035.e5. [PMID: 31091443 DOI: 10.1016/j.celrep.2019.04.074] [Citation(s) in RCA: 257] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 02/20/2019] [Accepted: 04/16/2019] [Indexed: 01/23/2023] Open
Abstract
The necessity of mesenchymal stromal cells, called fibroadipogenic progenitors (FAPs), in skeletal muscle regeneration and maintenance remains unestablished. We report the generation of a PDGFRαCreER knockin mouse model that provides a specific means of labeling and targeting FAPs. Depletion of FAPs using Cre-dependent diphtheria toxin expression results in loss of expansion of muscle stem cells (MuSCs) and CD45+ hematopoietic cells after injury and impaired skeletal muscle regeneration. Furthermore, FAP-depleted mice under homeostatic conditions exhibit muscle atrophy and loss of MuSCs, revealing that FAPs are required for the maintenance of both skeletal muscle and the MuSC pool. We also report that local tamoxifen metabolite delivery to target CreER activity in a single muscle, removing potentially confounding systemic effects of ablating PDGFRα+ cells distantly, also causes muscle atrophy. These data establish a critical role of FAPs in skeletal muscle regeneration and maintenance.
Collapse
Affiliation(s)
- Michael N Wosczyna
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Colin T Konishi
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Edgar E Perez Carbajal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Theodore T Wang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rachel A Walsh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Qiang Gan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mark W Wagner
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| |
Collapse
|
78
|
Rajendiran S, Smith-Berdan S, Kunz L, Risolino M, Selleri L, Schroeder T, Forsberg EC. Ubiquitous overexpression of CXCL12 confers radiation protection and enhances mobilization of hematopoietic stem and progenitor cells. Stem Cells 2020; 38:1159-1174. [PMID: 32442338 DOI: 10.1002/stem.3205] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
C-X-C motif chemokine ligand 12 (CXCL12; aka SDF1α) is a major regulator of a number of cellular systems, including hematopoiesis, where it influences hematopoietic cell trafficking, proliferation, and survival during homeostasis and upon stress and disease. A variety of constitutive, temporal, ubiquitous, and cell-specific loss-of-function models have documented the functional consequences on hematopoiesis upon deletion of Cxcl12. Here, in contrast to loss-of-function experiments, we implemented a gain-of-function approach by generating a doxycycline-inducible transgenic mouse model that enables spatial and temporal overexpression of Cxcl12. We demonstrated that ubiquitous CXCL12 overexpression led to an increase in multipotent progenitors in the bone marrow and spleen. The CXCL12+ mice displayed reduced reconstitution potential as either donors or recipients in transplantation experiments. Additionally, we discovered that Cxcl12 overexpression improved hematopoietic stem and progenitor cell mobilization into the blood, and conferred radioprotection by promoting quiescence. Thus, this new CXCL12+ mouse model provided new insights into major facets of hematopoiesis and serves as a versatile resource for studying CXCL12 function in a variety of contexts.
Collapse
Affiliation(s)
- Smrithi Rajendiran
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| | - Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| | - Leo Kunz
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| | - Maurizio Risolino
- Program in Craniofacial Biology, Institute of Human Genetics, Eli and Edyth Broad Center of Regeneration Medicine and Stem Cell Research, Departments of Orofacial Sciences and Anatomy, University of California, San Francisco, California, USA
| | - Licia Selleri
- Program in Craniofacial Biology, Institute of Human Genetics, Eli and Edyth Broad Center of Regeneration Medicine and Stem Cell Research, Departments of Orofacial Sciences and Anatomy, University of California, San Francisco, California, USA
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, Basel, Switzerland
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California, USA
| |
Collapse
|
79
|
Delgado RN, Mansky B, Ahanger SH, Lu C, Andersen RE, Dou Y, Alvarez-Buylla A, Lim DA. Maintenance of neural stem cell positional identity by mixed-lineage leukemia 1. Science 2020; 368:48-53. [PMID: 32241942 DOI: 10.1126/science.aba5960] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 03/09/2020] [Indexed: 12/22/2022]
Abstract
Neural stem cells (NSCs) in the developing and postnatal brain have distinct positional identities that dictate the types of neurons they generate. Although morphogens initially establish NSC positional identity in the neural tube, it is unclear how such regional differences are maintained as the forebrain grows much larger and more anatomically complex. We found that the maintenance of NSC positional identity in the murine brain requires a mixed-lineage leukemia 1 (Mll1)-dependent epigenetic memory system. After establishment by sonic hedgehog, ventral NSC identity became independent of this morphogen. Even transient MLL1 inhibition caused a durable loss of ventral identity, resulting in the generation of neurons with the characteristics of dorsal NSCs in vivo. Thus, spatial information provided by morphogens can be transitioned to epigenetic mechanisms that maintain regionally distinct developmental programs in the forebrain.
Collapse
Affiliation(s)
- Ryan N Delgado
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94143, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Benjamin Mansky
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sajad Hamid Ahanger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| | - Changqing Lu
- Department of Human Anatomy, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, P.R. China
| | - Rebecca E Andersen
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,Developmental and Stem Cell Biology Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yali Dou
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Arturo Alvarez-Buylla
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Daniel A Lim
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94143, USA. .,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA.,San Francisco Veterans Affairs Medical Center, San Francisco, CA 94121, USA
| |
Collapse
|
80
|
Liu S, Lockhart JR, Fontenard S, Berlett M, Ryan TM. Mapping the Chromosomal Insertion Site of the GFP Transgene of UBC-GFP Mice to the MHC Locus. THE JOURNAL OF IMMUNOLOGY 2020; 204:1982-1987. [PMID: 32122998 DOI: 10.4049/jimmunol.1901338] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/01/2020] [Indexed: 12/16/2022]
Abstract
GFP is frequently used as a marker for tracking donor cells adoptively transplanted into recipient animals. The human ubiquitin C promoter (UBC)-driven-GFP transgenic mouse is a commonly used source of donor cells for this purpose. This mouse was initially generated in the C57BL/6 inbred strain and has been backcrossed into the BALB/cBy strain for over 11 generations. Both the C57BL/6 inbred and BALB/cBy congenic UBC-GFP lines are commercially available and have been widely distributed. These UBC-GFP lines can be a convenient resource for tracking donor cells in both syngenic MHC-matched and in allogenic MHC-mismatched studies as C57BL/6 (H-2b) and BALB/cBy (H-2d) have disparate MHC haplotypes. In this report, we surprisingly discover that the UBC-GFP BALB/cBy congenic mice still retain the H-2b MHC haplotype of their original C57BL/6 founder, suggesting that the UBC-GFP transgene integration site is closely linked to the MHC locus on chromosome 17. Using linear amplification-mediated PCR, we successfully map the UBC-GFP transgene to the MHC locus. This study highlights the importance and urgency of mapping the transgene integration site of transgenic mouse strains used in biomedical research. Furthermore, this study raises the possibility of alternative interpretations of previous studies using congenic UBC-GFP mice and focuses attention on the necessity for rigor and reproducibility in scientific research.
Collapse
Affiliation(s)
- Shanrun Liu
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Jonathan R Lockhart
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Suean Fontenard
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Mike Berlett
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Thomas M Ryan
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
81
|
Galeano Niño JL, Tay SS, Tearle JLE, Xie J, Govendir MA, Kempe D, Mazalo J, Drew AP, Colakoglu F, Kummerfeld SK, Proud CG, Biro M. The Lifeact-EGFP mouse is a translationally controlled fluorescent reporter of T cell activation. J Cell Sci 2020; 133:jcs238014. [PMID: 32041902 DOI: 10.1242/jcs.238014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
It has become increasingly evident that T cell functions are subject to translational control in addition to transcriptional regulation. Here, by using live imaging of CD8+ T cells isolated from the Lifeact-EGFP mouse, we show that T cells exhibit a gain in fluorescence intensity following engagement of cognate tumour target cells. The GFP signal increase is governed by Erk1/2-dependent distal T cell receptor (TCR) signalling and its magnitude correlates with IFN-γ and TNF-α production, which are hallmarks of T cell activation. Enhanced fluorescence was due to increased translation of Lifeact-EGFP protein, without an associated increase in its mRNA. Activation-induced gains in fluorescence were also observed in naïve and CD4+ T cells from the Lifeact-EGFP reporter, and were readily detected by both flow cytometry and live cell microscopy. This unique, translationally controlled reporter of effector T cell activation simultaneously enables tracking of cell morphology, F-actin dynamics and activation state in individual migrating T cells. It is a valuable addition to the limited number of reporters of T cell dynamics and activation, and opens the door to studies of translational activity and heterogeneities in functional T cell responses in situ.
Collapse
Affiliation(s)
- Jorge Luis Galeano Niño
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Szun S Tay
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jacqueline L E Tearle
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jianling Xie
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
| | - Matt A Govendir
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Daryan Kempe
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jessica Mazalo
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Alexander P Drew
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Feyza Colakoglu
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Sarah K Kummerfeld
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Christopher G Proud
- Lifelong Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, SA 5000, Australia
- School of Biological Sciences, University of Adelaide, Frome Road, Adelaide
| | - Maté Biro
- EMBL Australia, Single Molecule Science Node, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
82
|
Kathiriya JJ, Brumwell AN, Jackson JR, Tang X, Chapman HA. Distinct Airway Epithelial Stem Cells Hide among Club Cells but Mobilize to Promote Alveolar Regeneration. Cell Stem Cell 2020; 26:346-358.e4. [PMID: 31978363 DOI: 10.1016/j.stem.2019.12.014] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 01/01/2023]
Abstract
Lung injury activates specialized adult epithelial progenitors to regenerate the epithelium. Depending on the extent of injury, both remaining alveolar type II cells (AEC2s) and distal airway stem/progenitors mobilize to cover denuded alveoli and restore normal barriers. The major source of airway stem/progenitors other than basal-like cells remains uncertain. Here, we define a distinct subpopulation (∼5%) of club-like lineage-negative epithelial progenitors (LNEPs) marked by high H2-K1 expression critical for alveolar repair. Quiescent H2-K1high cells account for virtually all in vitro regenerative activity of airway lineages. After bleomycin injury, H2-K1 cells expand and differentiate in vivo to alveolar lineages. However, injured H2-K1 cells eventually develop impaired self-renewal with features of senescence, limiting complete repair. Normal H2-K1high cells transplanted into injured lungs differentiate into alveolar cells and rescue lung function. These findings indicate that small subpopulations of specialized stem/progenitors are required for effective lung regeneration and are a potential therapeutic adjunct after major lung injury.
Collapse
Affiliation(s)
- Jaymin J Kathiriya
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA
| | - Alexis N Brumwell
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA
| | - Julia R Jackson
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA
| | - Xiaodan Tang
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA; Department of Pulmonary Disease, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Harold A Chapman
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, San Francisco, CA 94143, USA.
| |
Collapse
|
83
|
β4GALT1 controls β1 integrin function to govern thrombopoiesis and hematopoietic stem cell homeostasis. Nat Commun 2020; 11:356. [PMID: 31953383 PMCID: PMC6968998 DOI: 10.1038/s41467-019-14178-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 12/13/2019] [Indexed: 12/26/2022] Open
Abstract
Glycosylation is critical to megakaryocyte (MK) and thrombopoiesis in the context of gene mutations that affect sialylation and galactosylation. Here, we identify the conserved B4galt1 gene as a critical regulator of thrombopoiesis in MKs. β4GalT1 deficiency increases the number of fully differentiated MKs. However, the resulting lack of glycosylation enhances β1 integrin signaling leading to dysplastic MKs with severely impaired demarcation system formation and thrombopoiesis. Platelets lacking β4GalT1 adhere avidly to β1 integrin ligands laminin, fibronectin, and collagen, while other platelet functions are normal. Impaired thrombopoiesis leads to increased plasma thrombopoietin (TPO) levels and perturbed hematopoietic stem cells (HSCs). Remarkably, β1 integrin deletion, specifically in MKs, restores thrombopoiesis. TPO and CXCL12 regulate β4GalT1 in the MK lineage. Thus, our findings establish a non-redundant role for β4GalT1 in the regulation of β1 integrin function and signaling during thrombopoiesis. Defective thrombopoiesis and lack of β4GalT1 further affect HSC homeostasis.
Collapse
|
84
|
Haghayegh Jahromi N, Marchetti L, Moalli F, Duc D, Basso C, Tardent H, Kaba E, Deutsch U, Pot C, Sallusto F, Stein JV, Engelhardt B. Intercellular Adhesion Molecule-1 (ICAM-1) and ICAM-2 Differentially Contribute to Peripheral Activation and CNS Entry of Autoaggressive Th1 and Th17 Cells in Experimental Autoimmune Encephalomyelitis. Front Immunol 2020; 10:3056. [PMID: 31993059 PMCID: PMC6970977 DOI: 10.3389/fimmu.2019.03056] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/16/2019] [Indexed: 12/22/2022] Open
Abstract
In experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), myelin-specific T cells are activated in the periphery and differentiate in T helper (Th) 1 and Th17 effector cells, which cross the blood-brain barrier (BBB) to reach the central nervous system (CNS), where they induce neuroinflammation. Here, we explored the role of intercellular adhesion molecule-1 (ICAM-1) and ICAM-2 in the activation of naïve myelin-specific T cells and in the subsequent migration of differentiated encephalitogenic Th1 and Th17 cells across the BBB in vitro and in vivo. While on antigen-presenting cells ICAM-1, but not ICAM-2 was required for the activation of naïve CD4+ T cells, endothelial ICAM-1 and ICAM-2 mediated both Th1 and Th17 cell migration across the BBB. ICAM-1/-2-deficient mice developed ameliorated typical and atypical EAE transferred by encephalitogenic Th1 and Th17 cells, respectively. Our study underscores important yet cell-specific contributions for ICAM-1 and ICAM-2 in EAE pathogenesis.
Collapse
Affiliation(s)
| | - Luca Marchetti
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Federica Moalli
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Donovan Duc
- Laboratories of Neuroimmunology, Division of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, Epalinges, Switzerland
| | - Camilla Basso
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Heidi Tardent
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Elisa Kaba
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Urban Deutsch
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Division of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital, University of Lausanne, Epalinges, Switzerland
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland.,Department of Biology, Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Jens V Stein
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | |
Collapse
|
85
|
Faltusová K, Báječný M, Heizer T, Páral P, Nečas E. T-lymphopoiesis is Severely Compromised in Ubiquitin-Green Fluorescent Protein Transgenic Mice. Folia Biol (Praha) 2020; 66:47-59. [PMID: 32851834 DOI: 10.14712/fb2020066020047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Tagging cells of experimental organisms with genetic markers is commonly used in biomedical research. Insertion of artificial gene constructs can be highly beneficial for research as long as this tagging is functionally neutral and does not alter the tissue function. The transgenic UBC-GFP mouse has been recently found to be questionable in this respect, due to a latent stem cell defect compromising its lymphopoiesis and significantly influencing the results of competitive transplantation assays. In this study, we show that the stem cell defect present in UBC-GFP mice negatively affects T-lymphopoiesis significantly more than B-lymphopoiesis. The production of granulocytes is not negatively affected. The defect in T-lymphopoiesis causes a low total number of white blood cells in the peripheral blood of UBC-GFP mice which, together with the lower lymphoid/myeloid ratio in nucleated blood cells, is the only abnormal phenotype in untreated UBCGFP mice to have been found to date. The defective lymphopoiesis in UBC-GFP mice can be repaired by transplantation of congenic wild-type bone marrow cells, which then compensate for the insufficient production of T cells. Interestingly, the wild-type branch of haematopoiesis in chimaeric UBC-GFP/wild-type mice was more active in lymphopoiesis, and particularly towards production of T cells, compared to the lymphopoiesis in normal wild-type donors.
Collapse
Affiliation(s)
- K Faltusová
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - M Báječný
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - T Heizer
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - P Páral
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - E Nečas
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
86
|
Nakamura-Ishizu A, Matsumura T, Stumpf PS, Umemoto T, Takizawa H, Takihara Y, O'Neil A, Majeed ABBA, MacArthur BD, Suda T. Thrombopoietin Metabolically Primes Hematopoietic Stem Cells to Megakaryocyte-Lineage Differentiation. Cell Rep 2019; 25:1772-1785.e6. [PMID: 30428347 DOI: 10.1016/j.celrep.2018.10.059] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/07/2018] [Accepted: 10/15/2018] [Indexed: 02/07/2023] Open
Abstract
During acute myelosuppression or thrombocytopenia, bone marrow (BM) hematopoietic cells respond rapidly to replenish peripheral blood platelets. While the cytokine thrombopoietin (Thpo) both regulates platelet production and maintains HSC potential, whether Thpo controls megakaryocyte (Mk)-lineage differentiation of HSCs is unclear. Here, we show that Thpo rapidly upregulates mitochondrial activity in HSCs, an activity accompanied by differentiation to an Mk lineage. Moreover, in unperturbed hematopoiesis, HSCs with high mitochondrial activity exhibit Mk-lineage differentiation in vitro and myeloid lineage-biased reconstitution in vivo. Furthermore, Thpo skewed HSCs to express the tetraspanin CD9, a pattern correlated with mitochondrial activity. Mitochondria-active HSCs are resistant to apoptosis and oxidative stress upon Thpo stimulation. Thpo-regulated mitochondrial activity associated with mitochondrial translocation of STAT3 phosphorylated at serine 727. Overall, we report an important role for Thpo in regulating rapid Mk-lineage commitment. Thpo-dependent changes in mitochondrial metabolism prime HSCs to undergo direct differentiation to an Mk lineage.
Collapse
Affiliation(s)
- Ayako Nakamura-Ishizu
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan.
| | - Takayoshi Matsumura
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore
| | - Patrick S Stumpf
- Centre for Human Development Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Terumasa Umemoto
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan
| | - Hitoshi Takizawa
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan
| | - Yuji Takihara
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore
| | - Aled O'Neil
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore
| | | | - Ben D MacArthur
- International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan; Mathematical Sciences, University of Southampton, Southampton SO17 1BJ, UK; Centre for Human Development Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
| | - Toshio Suda
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, MD6, 117599 Singapore, Singapore; International Research Center for Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto City 860-0811, Japan.
| |
Collapse
|
87
|
Convergence between Microglia and Peripheral Macrophages Phenotype during Development and Neuroinflammation. J Neurosci 2019; 40:784-795. [PMID: 31818979 DOI: 10.1523/jneurosci.1523-19.2019] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/30/2022] Open
Abstract
Differently from other myeloid cells, microglia derive exclusively from precursors originating within the yolk sac and migrate to the CNS under development, without any contribution from fetal liver or postnatal hematopoiesis. Consistent with their unique ontology, microglia may express specific physiological markers, which have been partly described in recent years. Here we wondered whether profiles distinguishing microglia from peripheral macrophages vary with age and under pathology. To this goal, we profiled transcriptomes of microglia throughout the lifespan and included a parallel comparison with peripheral macrophages under physiological and neuroinflammatory settings using age- and sex-matched wild-type and bone marrow chimera mouse models. This comprehensive approach demonstrated that the phenotypic differentiation between microglia and peripheral macrophages is age-dependent and that peripheral macrophages do express some of the most commonly described microglia-specific markers early during development, such as Fcrls, P2ry12, Tmem119, and Trem2. Further, during chronic neuroinflammation CNS-infiltrating macrophages and not peripheral myeloid cells acquire microglial markers, indicating that the CNS niche may instruct peripheral myeloid cells to gain the phenotype and, presumably, the function of the microglia cell. In conclusion, our data provide further evidence about the plasticity of the myeloid cell and suggest caution in the strict definition and application of microglia-specific markers.SIGNIFICANCE STATEMENT Understanding the respective role of microglia and infiltrating monocytes in neuroinflammatory conditions has recently seemed possible by the identification of a specific microglia signature. Here instead we provide evidence that peripheral macrophages may express some of the most commonly described microglia markers at some developmental stages or pathological conditions, in particular during chronic neuroinflammation. Further, our data support the hypothesis about phenotypic plasticity and convergence among distinct myeloid cells so that they may act as a functional unit rather than as different entities, boosting their mutual functions in different phases of disease. This holds relevant implications in the view of the growing use of myeloid cell therapies to treat brain disease in humans.
Collapse
|
88
|
Lopez CK, Noguera E, Stavropoulou V, Robert E, Aid Z, Ballerini P, Bilhou-Nabera C, Lapillonne H, Boudia F, Thirant C, Fagnan A, Arcangeli ML, Kinston SJ, Diop M, Job B, Lecluse Y, Brunet E, Babin L, Villeval JL, Delabesse E, Peters AHFM, Vainchenker W, Gaudry M, Masetti R, Locatelli F, Malinge S, Nerlov C, Droin N, Lobry C, Godin I, Bernard OA, Göttgens B, Petit A, Pflumio F, Schwaller J, Mercher T. Ontogenic Changes in Hematopoietic Hierarchy Determine Pediatric Specificity and Disease Phenotype in Fusion Oncogene-Driven Myeloid Leukemia. Cancer Discov 2019; 9:1736-1753. [PMID: 31662298 DOI: 10.1158/2159-8290.cd-18-1463] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 08/05/2019] [Accepted: 09/23/2019] [Indexed: 01/18/2023]
Abstract
Fusion oncogenes are prevalent in several pediatric cancers, yet little is known about the specific associations between age and phenotype. We observed that fusion oncogenes, such as ETO2-GLIS2, are associated with acute megakaryoblastic or other myeloid leukemia subtypes in an age-dependent manner. Analysis of a novel inducible transgenic mouse model showed that ETO2-GLIS2 expression in fetal hematopoietic stem cells induced rapid megakaryoblastic leukemia whereas expression in adult bone marrow hematopoietic stem cells resulted in a shift toward myeloid transformation with a strikingly delayed in vivo leukemogenic potential. Chromatin accessibility and single-cell transcriptome analyses indicate ontogeny-dependent intrinsic and ETO2-GLIS2-induced differences in the activities of key transcription factors, including ERG, SPI1, GATA1, and CEBPA. Importantly, switching off the fusion oncogene restored terminal differentiation of the leukemic blasts. Together, these data show that aggressiveness and phenotypes in pediatric acute myeloid leukemia result from an ontogeny-related differential susceptibility to transformation by fusion oncogenes. SIGNIFICANCE: This work demonstrates that the clinical phenotype of pediatric acute myeloid leukemia is determined by ontogeny-dependent susceptibility for transformation by oncogenic fusion genes. The phenotype is maintained by potentially reversible alteration of key transcription factors, indicating that targeting of the fusions may overcome the differentiation blockage and revert the leukemic state.See related commentary by Cruz Hernandez and Vyas, p. 1653.This article is highlighted in the In This Issue feature, p. 1631.
Collapse
Affiliation(s)
- Cécile K Lopez
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Esteve Noguera
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Vaia Stavropoulou
- University Children's Hospital Beider Basel (UKBB) and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Elie Robert
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Zakia Aid
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | | | | | | | - Fabien Boudia
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
- Université Paris Diderot, Paris, France
| | - Cécile Thirant
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Alexandre Fagnan
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
- Université Paris Diderot, Paris, France
| | - Marie-Laure Arcangeli
- Unité Mixte de Recherche 967 INSERM, CEA/DRF/IBFJ/IRCM/LSHL, Université Paris-Diderot-Université Paris-Sud, Equipe labellisée Association Recherche Contre le Cancer, Fontenay-aux-roses, France
| | - Sarah J Kinston
- Wellcome and MRC Cambridge Stem Cell Institute and the Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | - Erika Brunet
- Genome Dynamics in the Immune System Laboratory, Institut Imagine, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Loélia Babin
- Genome Dynamics in the Immune System Laboratory, Institut Imagine, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Jean Luc Villeval
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Eric Delabesse
- INSERM U1037, Team 16, Center of Research of Cancerology of Toulouse, Hematology Laboratory, IUCT-Oncopole, France
| | - Antoine H F M Peters
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
- Faculty of Sciences, University of Basel, Basel, Switzerland
| | - William Vainchenker
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Muriel Gaudry
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Riccardo Masetti
- Department of Pediatrics, "Lalla Seràgnoli," Hematology-Oncology Unit, Sant'Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Franco Locatelli
- Department of Pediatrics, Sapienza, University of Rome, Rome, Italy
- Hematology-Oncology-IRCCS Ospedale Bambino Gesù, Rome, Italy
| | - Sébastien Malinge
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Claus Nerlov
- MRC Molecular Hematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | | | - Isabelle Godin
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Olivier A Bernard
- INSERM U1170, Gustave Roussy, Villejuif, France
- Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Berthold Göttgens
- Wellcome and MRC Cambridge Stem Cell Institute and the Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | | | - Françoise Pflumio
- Unité Mixte de Recherche 967 INSERM, CEA/DRF/IBFJ/IRCM/LSHL, Université Paris-Diderot-Université Paris-Sud, Equipe labellisée Association Recherche Contre le Cancer, Fontenay-aux-roses, France
| | - Juerg Schwaller
- University Children's Hospital Beider Basel (UKBB) and Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Thomas Mercher
- INSERM U1170, Gustave Roussy, Villejuif, France.
- Gustave Roussy, Villejuif, France
- Equipe labellisée Ligue Nationale Contre le Cancer, Paris, France
- Université Paris Diderot, Paris, France
| |
Collapse
|
89
|
Pizzagalli DU, Latino I, Pulfer A, Palomino-Segura M, Virgilio T, Farsakoglu Y, Krause R, Gonzalez SF. Characterization of the Dynamic Behavior of Neutrophils Following Influenza Vaccination. Front Immunol 2019; 10:2621. [PMID: 31824481 PMCID: PMC6881817 DOI: 10.3389/fimmu.2019.02621] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/22/2019] [Indexed: 12/24/2022] Open
Abstract
Neutrophils are amongst the first cells to respond to inflammation and infection. Although they play a key role in limiting the dissemination of pathogens, the study of their dynamic behavior in immune organs remains elusive. In this work, we characterized in vivo the dynamic behavior of neutrophils in the mouse popliteal lymph node (PLN) after influenza vaccination with UV-inactivated virus. To achieve this, we used an image-based systems biology approach to detect the motility patterns of neutrophils and to associate them to distinct actions. We described a prominent and rapid recruitment of neutrophils to the PLN following vaccination, which was dependent on the secretion of the chemokine CXCL1 and the alarmin molecule IL-1α. In addition, we observed that the initial recruitment occurred mainly via high endothelial venules located in the paracortical and interfollicular regions of the PLN. The analysis of the spatial-temporal patterns of neutrophil migration demonstrated that, in the initial stage, the majority of neutrophils displayed a patrolling behavior, followed by the formation of swarms in the subcapsular sinus of the PLN, which were associated with macrophages in this compartment. Finally, we observed using multiple imaging techniques, that neutrophils phagocytize and transport influenza virus particles. These processes might have important implications in the capacity of these cells to present viral antigens.
Collapse
Affiliation(s)
- Diego Ulisse Pizzagalli
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Computational Science, Università della Svizzera italiana, Lugano, Switzerland
| | - Irene Latino
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Alain Pulfer
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Miguel Palomino-Segura
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Tommaso Virgilio
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | | | - Rolf Krause
- Institute of Computational Science, Università della Svizzera italiana, Lugano, Switzerland
| | - Santiago F. Gonzalez
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| |
Collapse
|
90
|
TraFo-CRISPR: Enhanced Genome Engineering by Transient Foamy Virus Vector-Mediated Delivery of CRISPR/Cas9 Components. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:708-726. [PMID: 31726388 PMCID: PMC6859288 DOI: 10.1016/j.omtn.2019.10.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 12/26/2022]
Abstract
The adaptation of CRISPR/Cas technology for use in mammals has revolutionized genome engineering. In particular with regard to clinical application, efficient expression of Cas9 within a narrow time frame is highly desirable to minimize the accumulation of off-target editing. We developed an effective, aptamer-independent retroviral delivery system for Cas9 mRNAs that takes advantage of a unique foamy virus (FV) capability: the efficient encapsidation and transfer of non-viral RNAs. This enabled us to create a FV vector toolbox for efficient, transient delivery (TraFo) of CRISPR/Cas9 components into different target tissues. Co-delivery of Cas9 mRNA by TraFo-Cas9 vectors in combination with retroviral, integration-deficient single guide RNA (sgRNA) expression enhanced efficacy and specificity of gene-inactivation compared with CRISPR/Cas9 lentiviral vector systems. Furthermore, separate TraFo-Cas9 delivery allowed the optional inclusion of a repair matrix for efficient gene correction or tagging as well as the addition of fluorescent negative selection markers for easy identification of off-target editing or incorrect repair events. Thus, the TraFo CRISPR toolbox represents an interesting alternative technology for gene inactivation and gene editing.
Collapse
|
91
|
Peng Y, Liou B, Inskeep V, Blackwood R, Mayhew CN, Grabowski GA, Sun Y. Intravenous infusion of iPSC-derived neural precursor cells increases acid β-glucosidase function in the brain and lessens the neuronopathic phenotype in a mouse model of Gaucher disease. Hum Mol Genet 2019; 28:3406-3421. [PMID: 31373366 PMCID: PMC6891072 DOI: 10.1093/hmg/ddz184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 07/12/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023] Open
Abstract
Gaucher disease (GD) is caused by GBA1 mutations leading to functional deficiency of acid-β-glucosidase (GCase). No effective treatment is available for neuronopathic GD (nGD). A subclass of neural stem and precursor cells (NPCs) expresses VLA4 (integrin α4β1, very late antigen-4) that facilitates NPC entry into the brain following intravenous (IV) infusion. Here, the therapeutic potential of IV VLA4+NPCs was assessed for nGD using wild-type mouse green fluorescent protein (GFP)-positive multipotent induced pluripotent stem cell (iPSC)-derived VLA4+NPCs. VLA4+NPCs successfully engrafted in the nGD (4L;C*) mouse brain. GFP-positive cells differentiated into neurons, astrocytes and oligodendrocytes in the brainstem, midbrain and thalamus of the transplanted mice and significantly improved sensorimotor function and prolonged life span compared to vehicle-treated 4L;C* mice. VLA4+NPC transplantation significantly decreased levels of CD68 and glial fibrillary acidic protein, as well as TNFα mRNA levels in the brain, indicating reduced neuroinflammation. Furthermore, decreased Fluoro-Jade C and NeuroSilver staining suggested inhibition of neurodegeneration. VLA4+NPC-engrafted 4L;C* midbrains showed 35% increased GCase activity, reduced substrate [glucosylceramide (GC, -34%) and glucosylsphingosine (GS, -11%)] levels and improved mitochondrial oxygen consumption rates in comparison to vehicle-4L;C* mice. VLA4+NPC engraftment in 4L;C* brain also led to enhanced expression of neurotrophic factors that have roles in neuronal survival and the promotion of neurogenesis. This study provides evidence that iPSC-derived NPC transplantation has efficacy in an nGD mouse model and provides proof of concept for autologous NPC therapy in nGD.
Collapse
Affiliation(s)
- Yanyan Peng
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Benjamin Liou
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Venette Inskeep
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rachel Blackwood
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Christopher N Mayhew
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Gregory A Grabowski
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ying Sun
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
92
|
Mekala SR, Wörsdörfer P, Bauer J, Stoll O, Wagner N, Reeh L, Loew K, Eckner G, Kwok CK, Wischmeyer E, Dickinson ME, Schulze H, Stegner D, Benndorf RA, Edenhofer F, Pfeiffer V, Kuerten S, Frantz S, Ergün S. Generation of Cardiomyocytes From Vascular Adventitia-Resident Stem Cells. Circ Res 2019; 123:686-699. [PMID: 30355234 DOI: 10.1161/circresaha.117.312526] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
RATIONALE Regeneration of lost cardiomyocytes is a fundamental unresolved problem leading to heart failure. Despite several strategies developed from intensive studies performed in the past decades, endogenous regeneration of heart tissue is still limited and presents a big challenge that needs to be overcome to serve as a successful therapeutic option for myocardial infarction. OBJECTIVE One of the essential prerequisites for cardiac regeneration is the identification of endogenous cardiomyocyte progenitors and their niche that can be targeted by new therapeutic approaches. In this context, we hypothesized that the vascular wall, which was shown to harbor different types of stem and progenitor cells, might serve as a source for cardiac progenitors. METHODS AND RESULTS We describe generation of spontaneously beating mouse aortic wall-derived cardiomyocytes without any genetic manipulation. Using aortic wall-derived cells (AoCs) of WT (wild type), αMHC (α-myosin heavy chain), and Flk1 (fetal liver kinase 1)-reporter mice and magnetic bead-associated cell sorting sorting of Flk1+ AoCs from GFP (green fluorescent protein) mice, we identified Flk1+CD (cluster of differentiation) 34+Sca-1 (stem cell antigen-1)-CD44- AoCs as the population that gives rise to aortic wall-derived cardiomyocytes. This AoC subpopulation delivered also endothelial cells and macrophages with a particular accumulation within the aortic wall-derived cardiomyocyte containing colonies. In vivo, cardiomyocyte differentiation capacity was studied by implantation of fluorescently labeled AoCs into chick embryonic heart. These cells acquired cardiomyocyte-like phenotype as shown by αSRA (α-sarcomeric actinin) expression. Furthermore, coronary adventitial Flk1+ and CD34+ cells proliferated, migrated into the myocardium after mouse myocardial infarction, and expressed Isl-1+ (insulin gene enhancer protein-1) indicative of cardiovascular progenitor potential. CONCLUSIONS Our data suggest Flk1+CD34+ vascular adventitia-resident stem cells, including those of coronary adventitia, as a novel endogenous source for generating cardiomyocytes. This process is essentially supported by endothelial cells and macrophages. In summary, the therapeutic manipulation of coronary adventitia-resident cardiac stem and their supportive cells may open new avenues for promoting cardiac regeneration and repair after myocardial infarction and for preventing heart failure.
Collapse
Affiliation(s)
- Subba Rao Mekala
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Philipp Wörsdörfer
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Jochen Bauer
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Olga Stoll
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Nicole Wagner
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Laurens Reeh
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Kornelia Loew
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Georg Eckner
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Chee Keong Kwok
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Erhard Wischmeyer
- Institute of Physiology (E.W.).,University of Würzburg, Germany; Department of Psychiatry, Psychosomatics, and Psychotherapy, Center of Mental Health (E.W.)
| | - Mary Eleanor Dickinson
- University Hospital of Wuerzburg, Germany; Baylor College of Medicine, Houston, TX (M.E.D.)
| | | | | | - Ralf A Benndorf
- Department of Clinical Pharmacy and Pharmacotherapy, University of Halle-Wittenberg, Germany (R.A.B.)
| | - Frank Edenhofer
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Verena Pfeiffer
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Stefanie Kuerten
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| | - Stefan Frantz
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.).,Department of Internal Medicine I, ZIM (Zentrum für Innere Medizin) (S.F.)
| | - Süleyman Ergün
- From the Institute of Anatomy and Cell Biology II (S.R.M., P.W., J.B., O.S., N.W., L.R., K.L., G.E., C.K.K., F.E., V.P., S.K., S.E.)
| |
Collapse
|
93
|
Casu C, Chessa R, Liu A, Gupta R, Drakesmith H, Fleming R, Ginzburg YZ, MacDonald B, Rivella S. Minihepcidins improve ineffective erythropoiesis and splenomegaly in a new mouse model of adult β-thalassemia major. Haematologica 2019; 105:1835-1844. [PMID: 31582543 PMCID: PMC7327634 DOI: 10.3324/haematol.2018.212589] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 09/26/2019] [Indexed: 01/11/2023] Open
Abstract
Minihepcidins are hepcidin agonists that have been previously shown to reverse iron overload and improve erythropoiesis in mice affected by non-transfusion-dependent thalassemia. Given the extreme anemia that occurred with the previous model of transfusion-dependent thalassemia, that model was inadequate for investigating whether minihepcidins can improve red blood cell quality, lifespan and ineffective erythropoiesis. To overcome this limitation, we generated a new murine model of transfusion-dependent thalassemia with severe anemia and splenomegaly, but sufficient red cells and hemoglobin production to test the effect of minihepcidins. Furthermore, this new model demonstrates cardiac iron overload for the first time. In the absence of transfusions, minihepcidins improved red blood cell morphology and lifespan as well as ineffective erythropoiesis. Administration of a minihepcidin in combination with chronic red blood cell transfusion further improved the ineffective erythropoiesis and splenomegaly and reversed cardiac iron overload. These studies indicate that drugs such as minihepcidins have therapeutic potential for patients with transfusion-dependent thalassemia.
Collapse
Affiliation(s)
- Carla Casu
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Roberta Chessa
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Alison Liu
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Ritama Gupta
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Robert Fleming
- Department of Pediatrics, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Yelena Z Ginzburg
- Division of Hematology and Medical Oncology, Tisch Cancer Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, The Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA
| |
Collapse
|
94
|
Ficht X, Ruef N, Stolp B, Samson GPB, Moalli F, Page N, Merkler D, Nichols BJ, Diz-Muñoz A, Legler DF, Niggli V, Stein JV. In Vivo Function of the Lipid Raft Protein Flotillin-1 during CD8 + T Cell-Mediated Host Surveillance. THE JOURNAL OF IMMUNOLOGY 2019; 203:2377-2387. [PMID: 31548330 DOI: 10.4049/jimmunol.1900075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/24/2019] [Indexed: 01/12/2023]
Abstract
Flotillin-1 (Flot1) is an evolutionary conserved, ubiquitously expressed lipid raft-associated scaffolding protein. Migration of Flot1-deficient neutrophils is impaired because of a decrease in myosin II-mediated contractility. Flot1 also accumulates in the uropod of polarized T cells, suggesting an analogous role in T cell migration. In this study, we analyzed morphology and migration parameters of murine wild-type and Flot1-/- CD8+ T cells using in vitro assays and intravital two-photon microscopy of lymphoid and nonlymphoid tissues. Flot1-/- CD8+ T cells displayed significant alterations in cell shape and motility parameters in vivo but showed comparable homing to lymphoid organs and intact in vitro migration to chemokines. Furthermore, their clonal expansion and infiltration into nonlymphoid tissues during primary and secondary antiviral immune responses was comparable to wild-type CD8+ T cells. Taken together, Flot1 plays a detectable but unexpectedly minor role for CD8+ T cell behavior under physiological conditions.
Collapse
Affiliation(s)
- Xenia Ficht
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Nora Ruef
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Bettina Stolp
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland.,Department for Infectious Diseases, Integrative Virology, Center for Integrative Infectious Disease Research, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Guerric P B Samson
- Biotechnology Institute Thurgau at the University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Federica Moalli
- Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland.,Scientific Institute for Research and Healthcare, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Nicolas Page
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Ben J Nichols
- Medical Research Council Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany; and
| | - Daniel F Legler
- Biotechnology Institute Thurgau at the University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Verena Niggli
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland;
| |
Collapse
|
95
|
Sivapatham S, Ficht X, Barreto de Albuquerque J, Page N, Merkler D, Stein JV. Initial Viral Inoculum Determines Kinapse-and Synapse-Like T Cell Motility in Reactive Lymph Nodes. Front Immunol 2019; 10:2086. [PMID: 31552034 PMCID: PMC6743022 DOI: 10.3389/fimmu.2019.02086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/19/2019] [Indexed: 11/13/2022] Open
Abstract
T cell activation in lymphoid tissue occurs through interactions with cognate peptide-major histocompatibility complex (pMHC)-presenting dendritic cells (DCs). Intravital imaging studies using ex vivo peptide-pulsed DCs have uncovered that cognate pMHC levels imprint a wide range of dynamic contacts between these two cell types. T cell-DC interactions vary between transient, "kinapse-like" contacts at low to moderate pMHC levels to immediate "synapse-like" arrest at DCs displaying high pMHC levels. To date, it remains unclear whether this pattern is recapitulated when the immune system faces a replicative agent, such as a virus, at low and high inoculum. Here, we locally administered low and high inoculum of lymphocytic choriomeningitis virus (LCMV) in mice to follow activation parameters of Ag-specific CD4+ and CD8+ T cells in draining lymph nodes (LNs) during the first 72 h post infection. We correlated these data with kinapse- and synapse-like motility patterns of Ag-specific T cells obtained by intravital imaging of draining LNs. Our data show that initial viral inoculum controls immediate synapse-like T cell arrest vs. continuous kinapse-like motility. This remains the case when the viral inoculum and thus the inflammatory microenvironment in draining LNs remains identical but cognate pMHC levels vary. Our data imply that the Ag-processing capacity of draining LNs is equipped to rapidly present high levels of cognate pMHC when antigenic material is abundant. Our findings further suggest that widespread T cell arrest during the first 72 h of an antimicrobial immune responses is not required to trigger proliferation. In sum, T cells adapt their scanning behavior according to available antigen levels during viral infections, with dynamic changes in motility occurring before detectable expression of early activation markers.
Collapse
Affiliation(s)
- Sujana Sivapatham
- Department of Oncology, Microbiology, and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Xenia Ficht
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | | | - Nicolas Page
- Division of Clinical Pathology, Department of Pathology and Immunology, University Hospital of Geneva, Geneva, Switzerland
| | - Doron Merkler
- Division of Clinical Pathology, Department of Pathology and Immunology, University Hospital of Geneva, Geneva, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology, and Immunology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
96
|
Mesenchyme-free expansion and transplantation of adult alveolar progenitor cells: steps toward cell-based regenerative therapies. NPJ Regen Med 2019; 4:17. [PMID: 31452939 PMCID: PMC6702233 DOI: 10.1038/s41536-019-0080-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022] Open
Abstract
Alveolar type-2 (AT2) cells are necessary for the lung’s regenerative response to epithelial insults such as influenza. However, current methods to expand these cells rely on mesenchymal co-culture, complicating the possibility of transplantation following acute injury. Here we developed several mesenchyme-free culture conditions that promote growth of murine AT2 organoids. Transplanting dissociated AT2 organoids into influenza-infected mice demonstrated that organoids engraft and either proliferate as AT2 cells or unexpectedly adopt a basal cell-like fate associated with maladaptive regeneration. Alternatively, transplanted primary AT2 cells also robustly engraft, maintaining their AT2 lineage while replenishing the alveolar type-1 (AT1) cell population in the epithelium. Importantly, pulse oximetry revealed significant increase in blood-oxygen saturation in primary AT2 recipients, indicating that transplanted cells also confer increased pulmonary function after influenza. We further demonstrated that both acid installation and bleomycin injury models are also amenable to AT2 transplantation. These studies provide additional methods to study AT2 progenitor potential, while serving as proof-of-principle for adoptive transfer of alveolar progenitors in potential therapeutic applications.
Collapse
|
97
|
Brownlie RJ, Wright D, Zamoyska R, Salmond RJ. Deletion of PTPN22 improves effector and memory CD8+ T cell responses to tumors. JCI Insight 2019; 5:127847. [PMID: 31335326 PMCID: PMC6777814 DOI: 10.1172/jci.insight.127847] [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] [Indexed: 12/17/2022] Open
Abstract
Adoptive T cell therapy (ACT) has been established as an efficacious methodology for the treatment of cancer. Identifying targets to enhance the antigen recognition, functional capacity and longevity of T cells has the potential to broaden the applicability of these approaches in the clinic. We previously reported that targeting expression of phosphotyrosine phosphatase, non-receptor type (PTPN) 22 in effector CD8+ T cells enhances the efficacy of ACT for tumor clearance in mice. In the current work, we demonstrate that, upon ACT, PTPN22-deficient effector CD8+ T cells afford greater protection against tumors expressing very low affinity antigen, but do not survive long-term in vivo. Persistence of CD8+ T cells following tumor clearance is improved by ACT of memory phenotype cells that have a distinct metabolic phenotype as compared to effector T cells. Importantly, PTPN22-deficient T cells have comparable capacity to form long-lived memory cells in vivo but enhanced anti-tumor activity in vivo and effector responses ex vivo. These findings provide key insight into the regulation of effector and memory T cell responses in vivo, and indicate that PTPN22 is a rationale target to improve ACT for cancer.
Collapse
Affiliation(s)
- Rebecca J Brownlie
- Leeds Institute of Medical Research at St. James's, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, United Kingdom
| | - David Wright
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Rose Zamoyska
- Institute of Immunology and Infection Research, University of Edinburgh, Ashworth Laboratories, Edinburgh, United Kingdom
| | - Robert J Salmond
- Leeds Institute of Medical Research at St. James's, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, United Kingdom
| |
Collapse
|
98
|
Mouse HSA+ immature cardiomyocytes persist in the adult heart and expand after ischemic injury. PLoS Biol 2019; 17:e3000335. [PMID: 31246945 PMCID: PMC6619826 DOI: 10.1371/journal.pbio.3000335] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 07/10/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
The assessment of the regenerative capacity of the heart has been compromised by the lack of surface signatures to characterize cardiomyocytes (CMs). Here, combined multiparametric surface marker analysis with single-cell transcriptional profiling and in vivo transplantation identify the main mouse fetal cardiac populations and their progenitors (PRGs). We found that CMs at different stages of differentiation coexist during development. We identified a population of immature heat stable antigen (HSA)/ cluster of differentiation 24 (CD24)+ CMs that persists throughout life and that, unlike other CM subsets, actively proliferates up to 1 week of age and engrafts cardiac tissue upon transplantation. In the adult heart, a discrete population of HSA/CD24+ CMs appears as mononucleated cells that increase in frequency after infarction. Our work identified cell surface signatures that allow the prospective isolation of CMs at all developmental stages and the detection of a subset of immature CMs throughout life that, although at reduced frequencies, are poised for activation in response to ischemic stimuli. This work opens new perspectives in the understanding and treatment of heart pathologies.
Collapse
|
99
|
Meijer RI, Hoevenaars FPM, Serné EH, Yudkin JS, Kokhuis TJA, Weijers EM, van Hinsbergh VWM, Smulders YM, Eringa EC. JNK2 in myeloid cells impairs insulin's vasodilator effects in muscle during early obesity development through perivascular adipose tissue dysfunction. Am J Physiol Heart Circ Physiol 2019; 317:H364-H374. [PMID: 31149833 DOI: 10.1152/ajpheart.00663.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Reduced vasodilator properties of insulin in obesity are caused by changes in perivascular adipose tissue and contribute to microvascular dysfunction in skeletal muscle. The causes of this dysfunction are unknown. The effects of a short-term Western diet on JNK2-expressing cells in perivascular adipose tissue (PVAT) on insulin-induced vasodilation and perfusion of skeletal muscle were assessed. In vivo, 2 wk of Western diet (WD) reduced whole body insulin sensitivity and insulin-stimulated muscle perfusion, determined using contrast ultrasonography during the hyperinsulinemic clamp. Ex vivo, WD triggered accumulation of PVAT in skeletal muscle and blunted its ability to facilitate insulin-induced vasodilation. Labeling of myeloid cells with green fluorescent protein identified bone marrow as a source of PVAT in muscle. To study whether JNK2-expressing inflammatory cells from bone marrow were involved, we transplanted JNK2-/- bone marrow to WT mice. Deletion of JNK2 in bone marrow rescued the vasodilator phenotype of PVAT during WD exposure. JNK2 deletion in myeloid cells prevented the WD-induced increase in F4/80 expression. Even though WD and JNK2 deletion resulted in specific changes in gene expression of PVAT; epididymal and subcutaneous adipose tissue; expression of tumor necrosis factor-α, interleukin-1β, interleukin-6, or protein inhibitor of STAT1 was not affected. In conclusion, short-term Western diet triggers infiltration of JNK2-positive myeloid cells into PVAT, resulting in PVAT dysfunction, nonclassical inflammation, and loss of insulin-induced vasodilatation in vivo and ex vivo.NEW & NOTEWORTHY We demonstrate that in the earliest phase of weight gain, changes in perivascular adipose tissue in muscle impair insulin-stimulated muscle perfusion. The hallmark of these changes is infiltration by inflammatory cells. Deletion of JNK2 from the bone marrow restores the function of perivascular adipose tissue to enhance insulin's vasodilator effects in muscle, showing that the bone marrow contributes to regulation of muscle perfusion.
Collapse
Affiliation(s)
- Rick I Meijer
- Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| | - Femke P M Hoevenaars
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| | - Erik H Serné
- Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| | - John S Yudkin
- Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands.,Department of Medicine, University College London, London, United Kingdom
| | - Tom J A Kokhuis
- Biomedical Engineering, Thorax Center, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ester M Weijers
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| | - Victor W M van Hinsbergh
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| | - Yvo M Smulders
- Department of Internal Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
100
|
Ludwicki MB, Li J, Stephens EA, Roberts RW, Koide S, Hammond PT, DeLisa MP. Broad-Spectrum Proteome Editing with an Engineered Bacterial Ubiquitin Ligase Mimic. ACS CENTRAL SCIENCE 2019; 5:852-866. [PMID: 31139721 PMCID: PMC6535771 DOI: 10.1021/acscentsci.9b00127] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Indexed: 05/03/2023]
Abstract
Manipulation of the ubiquitin-proteasome pathway to achieve targeted silencing of cellular proteins has emerged as a reliable and customizable strategy for remodeling the mammalian proteome. One such approach involves engineering bifunctional proteins called ubiquibodies that are comprised of a synthetic binding protein fused to an E3 ubiquitin ligase, thus enabling post-translational ubiquitination and degradation of a target protein independent of its function. Here, we have designed a panel of new ubiquibodies based on E3 ubiquitin ligase mimics from bacterial pathogens that are capable of effectively interfacing with the mammalian proteasomal degradation machinery for selective removal of proteins of interest. One of these, the Shigella flexneri effector protein IpaH9.8 fused to a fibronectin type III (FN3) monobody that specifically recognizes green fluorescent protein (GFP), was observed to potently eliminate GFP and its spectral derivatives as well as 15 different FP-tagged mammalian proteins that varied in size (27-179 kDa) and subcellular localization (cytoplasm, nucleus, membrane-associated, and transmembrane). To demonstrate therapeutically relevant delivery of ubiquibodies, we leveraged a bioinspired molecular assembly method whereby synthetic mRNA encoding the GFP-specific ubiquibody was coassembled with poly A binding proteins and packaged into nanosized complexes using biocompatible, structurally defined polypolypeptides bearing cationic amine side groups. The resulting nanoplexes delivered ubiquibody mRNA in a manner that caused efficient target depletion in cultured mammalian cells stably expressing GFP as well as in transgenic mice expressing GFP ubiquitously. Overall, our results suggest that IpaH9.8-based ubiquibodies are a highly modular proteome editing technology with the potential for pharmacologically modulating disease-causing proteins.
Collapse
Affiliation(s)
- Morgan B. Ludwicki
- Robert F. Smith
School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New
York 14853, United
States
| | - Jiahe Li
- Department of Chemical Engineering and Koch Institute for
Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Erin A. Stephens
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, New York 14853, United States
| | - Richard W. Roberts
- Department
of Chemistry, University of Southern California, Los Angeles, California 90089, United States
| | - Shohei Koide
- Perlmutter Cancer Center, New York University
Langone Medical Center, New York, New York 10016, United States
- Department of Biochemistry and Molecular
Pharmacology, New York University School
of Medicine, New York, New York 10016, United States
| | - Paula T. Hammond
- Department of Chemical Engineering and Koch Institute for
Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Matthew P. DeLisa
- Robert F. Smith
School of Chemical and Biomolecular Engineering, Cornell University, 120 Olin Hall, Ithaca, New
York 14853, United
States
- Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|