1
|
Zhang Y, Zhou J, Zhao J, Cheng X, Xing C. Chronic benzene exposure impairs the self-renewal capacity of HSPCs in C57BL/6 mice. Toxicol Res (Camb) 2025; 14:tfaf021. [PMID: 39968519 PMCID: PMC11831024 DOI: 10.1093/toxres/tfaf021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
In this study, we aimed to investigate the effects of chronic benzene exposure on the self-renewal capacity of C57BL/6 bone marrow HSPCs. Twenty-four male C57BL/6 mice were randomly divided into two groups: the control group and the benzene-exposed group. Mice in the benzene-exposed group inhaled 1,000 mg/m3 (308 ppm; conversion factor: 20 °C, 101 kPa, 1 ppm = 3.25 mg/m3) benzene for 32 weeks and the control group mice inhaled clean air. The peripheral blood hematological alterations were monitored every two weeks. Competitive bone marrow transplantation was performed to assess the self-renewal capacity of bone marrow HSPCs and the donor cell chimerism was quantified through flow cytometry. By the fourth week of benzene exposure, significant reductions in leukocytes, erythrocytes, and hemoglobin levels (P < 0.05) were observed, suggesting the development of benzene poisoning in mice. In the B6.SJL recipient mice, the chimerism ratio of bone marrow cells from C57BL/6 donors exposed to benzene for 20 weeks significantly decreased after transplantation (16 weeks post-transplant: 52.58% ± 17.38% in controls vs. 3.89% ± 1.96% in the benzene group, P < 0.05). Furthermore, the chimerism ratio in recipients of 32-week benzene-exposed donors approached zero by week 84 post-transplant, suggesting a loss of self-renewal capacity in bone marrow HSPCs due to benzene exposure. The study concludes that bone marrow suppression and the diminished self-renewal ability of C57BL/6 bone marrow HSPCs may lead to bone marrow failure in mice, contributing to the potential occurrence of MDS.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, 29 Nanwei Road, Xicheng District, Beijing 100050, China
| | - Jin Zhou
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, 29 Nanwei Road, Xicheng District, Beijing 100050, China
| | - Jianxin Zhao
- National Office for Maternal and Child Health Surveillance of China, West China Second University Hospital, Sichuan University, Sec. 3 No. 17 South Renmin Road, Wuhou District, Chengdu, Sichuan Province 610041, China
| | - Xiurong Cheng
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, 29 Nanwei Road, Xicheng District, Beijing 100050, China
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, 29 Nanwei Road, Xicheng District, Beijing 100050, China
| | - Caihong Xing
- State Key Laboratory of Trauma and Chemical Poisoning, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, 29 Nanwei Road, Xicheng District, Beijing 100050, China
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, 29 Nanwei Road, Xicheng District, Beijing 100050, China
| |
Collapse
|
2
|
Laubreton D, Djebali S, Angleraux C, Chain B, Dubois M, Henry F, Leverrier Y, Teixeira M, Markossian S, Marvel J. Generation of a C57BL/6J mouse strain expressing the CD45.1 epitope to improve hematopoietic stem cell engraftment and adoptive cell transfer experiments. Lab Anim (NY) 2023; 52:324-331. [PMID: 38017180 DOI: 10.1038/s41684-023-01275-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/05/2023] [Indexed: 11/30/2023]
Abstract
Adoptive cell transfer between genetically identical hosts relies on the use of a congenic marker to distinguish the donor cells from the host cells. CD45, a glycoprotein expressed by all hematopoietic cells, is one of the main congenic markers used because its two isoforms, CD45.1 and CD45.2, can be discriminated by flow cytometry. As a consequence, C57BL/6J (B6; CD45.2) and B6.SJL-Ptprca Pepcb/BoyJ (B6.SJL; CD45.1) mice are widely used in adoptive cell transfer experiments, under the presumption that they differ only at the CD45 (Ptprc) locus. However, recent studies have identified genetic variations between these congenic strains and have notably highlighted a differential expression of cathepsin E (CTSE). The B6.SJL mouse presents a number of functional differences in hematopoietic stem cell engraftment potential and immune cell numbers compared with the B6 mouse. In this study, we showed that B6 and B6.SJL mice also differ in their CD8+ T cell compartment and CD8+ T cell responses to viral infection. We identified Ctse as the most differentially expressed gene between CD8+ T cells of B6 and B6.SJL and demonstrated that the differences reported between these two mouse strains are not due to CTSE. Finally, using CRISPR-Cas9 genome editing, we generated a CD45.1-expressing B6 mouse by inserting one nucleotide mutation (A904G) leading to an amino acid change (K302E) in the Ptprc gene of the B6 mouse. We showed that this new B6-Ptprcem(K302E)Jmar/J mouse resolves the experimental biases reported between the B6 and B6.SJL mouse lines and should thus represent the new gold standard for adoptive cell transfer experiments in B6.
Collapse
Affiliation(s)
- Daphné Laubreton
- CIRI, INSERM U1111, Université Claude Bernard Lyon 1, CNRS UMR 5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Sophia Djebali
- CIRI, INSERM U1111, Université Claude Bernard Lyon 1, CNRS UMR 5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Céline Angleraux
- SFR BioSciences, Plateau de Biologie Expérimentale de la Souris (AniRA-PBES), Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UAR3444, INSERM US8, Lyon, France
| | - Benny Chain
- Division of Infection and Immunity, University College London, London, UK
| | - Maxence Dubois
- CIRI, INSERM U1111, Université Claude Bernard Lyon 1, CNRS UMR 5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Farida Henry
- SFR BioSciences, Plateau de Biologie Expérimentale de la Souris (AniRA-PBES), Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UAR3444, INSERM US8, Lyon, France
| | - Yann Leverrier
- CIRI, INSERM U1111, Université Claude Bernard Lyon 1, CNRS UMR 5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Marie Teixeira
- SFR BioSciences, Plateau de Biologie Expérimentale de la Souris (AniRA-PBES), Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, CNRS UAR3444, INSERM US8, Lyon, France
| | - Suzy Markossian
- Institut de Génomique Fonctionnelle de Lyon, INRAE USC 1370, CNRS UMR 5242, Ecole Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jacqueline Marvel
- CIRI, INSERM U1111, Université Claude Bernard Lyon 1, CNRS UMR 5308, École Normale Supérieure de Lyon, Université de Lyon, Lyon, France.
| |
Collapse
|
3
|
Kalari Kandy RR, Fan X, Cao X. CD45.1/CD45.2 Congenic Markers Induce a Selective Bias for CD8+ T Cells during Adoptive Lymphocyte Reconstitution in Lymphocytopenia Mice. Immunohorizons 2023; 7:755-759. [PMID: 37938184 PMCID: PMC10695411 DOI: 10.4049/immunohorizons.2300014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
CD45.1/CD45.2 congenic markers have been used to track hematopoietic lineage differentiation following hematopoietic stem and progenitor cell (HSPC) transplantation. However, several studies suggest that a bias exists in CD45.1 versus CD45.2 hematopoietic cell reconstitution from HSPCs. Meanwhile, no definitive comparison has been reported for mature immune cells as to whether the CD45.1/CD45.2 disparity can skew the immune cell response. In this study, using lymphocytopenia Rag1-/- CD45.2 mice as hosts, we assessed the reconstitution potential of CD45.1 versus CD45.2 lymphocytes following adoptive transfer of mature T and B cells. We have found a selective bias for CD8+ T cells in that CD45.1 cells showed significantly higher reconstitution compared with CD45.2 cells, whereas CD4+ T cells and CD19+ B cells showed equivalent reconstitution. These results suggest that CD45.1/CD45.2 markers may induce an alloreactive response or a survival bias specific to CD8+ T cells, and they therefore call for caution for using them as congenic markers in immunologic models.
Collapse
Affiliation(s)
- Rakhee Rathnam Kalari Kandy
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore School of Medicine, Baltimore, MD
| | - Xiaoxuan Fan
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore School of Medicine, Baltimore, MD
- Department of Microbiology and Immunology, University of Maryland Baltimore School of Medicine, Baltimore, MD
| | - Xuefang Cao
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore School of Medicine, Baltimore, MD
- Department of Microbiology and Immunology, University of Maryland Baltimore School of Medicine, Baltimore, MD
| |
Collapse
|
4
|
He C, Luo H, Coelho A, Liu M, Li Q, Xu J, Krämer A, Malin S, Yuan Z, Holmdahl R. NCF4 dependent intracellular reactive oxygen species regulate plasma cell formation. Redox Biol 2022; 56:102422. [PMID: 36095971 PMCID: PMC9482113 DOI: 10.1016/j.redox.2022.102422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/12/2022] [Accepted: 07/22/2022] [Indexed: 11/30/2022] Open
Abstract
Defective reactive oxygen species (ROS) production by genetically determined variants of the NADPH oxidase 2 (NOX2) complex component, NCF4, leads to enhanced production of autoantibodies to collagen type II (COL2) and severe collagen-induced arthritis (CIA) in mice. To further understand this process, we used mice harboring a mutation in the lipid endosomal membrane binding site (R58A) of NCF4 subunit. This mutation did not affect the extracellular ROS responses but showed instead decreased intracellular responses following B cell stimulation. Immunization with COL2 led to severe arthritis with increased antibody levels in Ncf458A mutated animals without significant effects on antigen presentation, autoreactive T cell activation and germinal center formation. Instead, plasma cell formation was enhanced and had altered CXCR3/CXCR4 expression. This B cell intrinsic effect was further confirmed with chimeric B cell transfer experiments and in vitro LPS or CD40L with anti-IgM stimulation. We conclude that NCF4 regulates the terminal differentiation of B cells to plasma cells through intracellular ROS. Ncf4R58A selectively affects intracellular ROS production after stimulation. Decreased intracellular ROS in B cell promotes plasma cell formation intrinsically. BCR stimulation induced NOX2 complex-ROS regulates CXCR3 expression on plasma cell.
Collapse
Affiliation(s)
- Chang He
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Huqiao Luo
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Ana Coelho
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Meng Liu
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; National Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Qijing Li
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China; Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; Department of Hematology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Jing Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Alexander Krämer
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Stephen Malin
- Department of Medicine Solna (MedS) Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Rikard Holmdahl
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; National Joint Engineering Research Center of Biodiagnostics and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
5
|
Roberto MP, Varano G, Vinas-Castells R, Holmes AB, Kumar R, Pasqualucci L, Farinha P, Scott DW, Dominguez-Sola D. Mutations in the transcription factor FOXO1 mimic positive selection signals to promote germinal center B cell expansion and lymphomagenesis. Immunity 2021; 54:1807-1824.e14. [PMID: 34380064 DOI: 10.1016/j.immuni.2021.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/26/2021] [Accepted: 07/13/2021] [Indexed: 12/27/2022]
Abstract
The transcription factor forkhead box O1 (FOXO1), which instructs the dark zone program to direct germinal center (GC) polarity, is typically inactivated by phosphatidylinositol 3-kinase (PI3K) signals. Here, we investigated how FOXO1 mutations targeting this regulatory axis in GC-derived B cell non-Hodgkin lymphomas (B-NHLs) contribute to lymphomagenesis. Examination of primary B-NHL tissues revealed that FOXO1 mutations and PI3K pathway activity were not directly correlated. Human B cell lines bearing FOXO1 mutations exhibited hyperactivation of PI3K and Stress-activated protein kinase (SAPK)/Jun amino-terminal kinase (JNK) signaling, and increased cell survival under stress conditions as a result of alterations in FOXO1 transcriptional affinities and activation of transcriptional programs characteristic of GC-positive selection. When modeled in mice, FOXO1 mutations conferred competitive advantage to B cells in response to key T-dependent immune signals, disrupting GC homeostasis. FOXO1 mutant transcriptional signatures were prevalent in human B-NHL and predicted poor clinical outcomes. Thus, rather than enforcing FOXO1 constitutive activity, FOXO1 mutations enable co-option of GC-positive selection programs during the pathogenesis of GC-derived lymphomas.
Collapse
Affiliation(s)
- Mark P Roberto
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gabriele Varano
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rosa Vinas-Castells
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Antony B Holmes
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Rahul Kumar
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA; Department of Pathology, Columbia University Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Pedro Farinha
- Center for Lymphoid Cancer, British Columbia Cancer, Vancouver, BC V5Z 1L3, Canada
| | - David W Scott
- Center for Lymphoid Cancer, British Columbia Cancer, Vancouver, BC V5Z 1L3, Canada
| | - David Dominguez-Sola
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
6
|
De novo generation of macrophage from placenta-derived hemogenic endothelium. Dev Cell 2021; 56:2121-2133.e6. [PMID: 34197725 DOI: 10.1016/j.devcel.2021.06.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 04/30/2021] [Accepted: 06/08/2021] [Indexed: 01/31/2023]
Abstract
Macrophages play pivotal roles in immunity, hematopoiesis, and tissue homeostasis. In mammals, macrophages have been shown to originate from yolk-sac-derived erythro-myeloid progenitors and aorta-gonad-mesonephros (AGM)-derived hematopoietic stem cells. However, whether macrophages can arise from other embryonic sites remains unclear. Here, using single-cell RNA sequencing, we profile the transcriptional landscape of mouse fetal placental hematopoiesis. We uncover and experimentally validate that a CD44+ subpopulation of placental endothelial cells (ECs) exhibits hemogenic potential. Importantly, lineage tracing using the newly generated Hoxa13 reporter line shows that Hoxa13-labeled ECs can produce placental macrophages, named Hofbauer cell (HBC)-like cells. Furthermore, we identify two subtypes of HBC-like cells, and cell-cell interaction analysis identifies their potential roles in angiogenesis and antigen presentation, separately. Our study provides a comprehensive understanding of placental hematopoiesis and highlights the placenta as a source of macrophages, which has important implications for both basic and translational research.
Collapse
|
7
|
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
|
8
|
Khalil M, Ray A, Dittel BN. Characterization of the Cell Surface Phenotype and Regulatory Activity of B-Cell IgD Low (BD L) Regulatory B Cells. Methods Mol Biol 2021; 2270:217-231. [PMID: 33479901 DOI: 10.1007/978-1-0716-1237-8_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
B-cell IgD Low (BDL) B cells have been shown to promote immunological tolerance by inducing proliferation of CD4+Foxp3+ T-regulatory cells (Treg) in a glucocorticoid-induced tumor necrosis factor receptor-related protein ligand (GITRL, Tnfsf18)-dependent manner. BDL cells constitute a small subset of splenic B lymphocytes that, in mice, are characterized by the B220+IgMintCD21intCD23+CD93-IgDlow/- cell surface expression profile. In this chapter, we show the flow cytometry gating strategy developed to identify and purify BDL. In addition, we describe an in vitro assay and two in vivo assays to assess BDL regulatory activity by quantitating Treg expansion/proliferation and indicate how they can be used in mouse models of disease. Collectively, these methods are useful to track and quantitate BDL and Treg numbers and assess their regulatory activity in inflammatory disease models.
Collapse
Affiliation(s)
- Mohamed Khalil
- Versiti Blood Research Institute, Milwaukee, WI, USA
- Molecular Biology Department, National Research Centre, Cairo, Egypt
| | - Avijit Ray
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | | |
Collapse
|
9
|
Huang QQ, Doyle R, Chen SY, Sheng Q, Misharin AV, Mao Q, Winter DR, Pope RM. Critical role of synovial tissue-resident macrophage niche in joint homeostasis and suppression of chronic inflammation. SCIENCE ADVANCES 2021; 7:7/2/eabd0515. [PMID: 33523968 PMCID: PMC7787490 DOI: 10.1126/sciadv.abd0515] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/17/2020] [Indexed: 05/30/2023]
Abstract
Little is known about the mechanisms regulating the transition of circulating monocytes into pro- or anti-inflammatory macrophages in chronic inflammation. Here, we took advantage of our novel mouse model of rheumatoid arthritis, in which Flip is deleted under the control of a CD11c promoter (HUPO mice). During synovial tissue homeostasis, both monocyte-derived F4/80int and self-renewing F4/80hi tissue-resident, macrophage populations were identified. However, in HUPO mice, decreased synovial tissue-resident macrophages preceded chronic arthritis, opened a niche permitting the influx of activated monocytes, with impaired ability to differentiate into F4/80hi tissue-resident macrophages. In contrast, Flip-replete monocytes entered the vacated niche and differentiated into tissue-resident macrophages, which suppressed arthritis. Genes important in macrophage tissue residency were reduced in HUPO F4/80hi macrophages and in leukocyte-rich rheumatoid arthritis synovial tissue monocytes. Our observations demonstrate that the macrophage tissue-resident niche is necessary for suppression of chronic inflammation and may contribute to the pathogenesis of rheumatoid arthritis.
Collapse
Affiliation(s)
- Qi-Quan Huang
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Renee Doyle
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Shang-Yang Chen
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qicong Sheng
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Alexander V Misharin
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Qinwen Mao
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Deborah R Winter
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Richard M Pope
- Department of Medicine, Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
10
|
Chisolm DA, Cheng W, Colburn SA, Silva-Sanchez A, Meza-Perez S, Randall TD, Weinmann AS. Defining Genetic Variation in Widely Used Congenic and Backcrossed Mouse Models Reveals Varied Regulation of Genes Important for Immune Responses. Immunity 2019; 51:155-168.e5. [PMID: 31248780 DOI: 10.1016/j.immuni.2019.05.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/24/2019] [Accepted: 05/15/2019] [Indexed: 12/27/2022]
Abstract
Genetic variation influences how the genome is interpreted in individuals and in mouse strains used to model immune responses. We developed approaches to utilize next-generation sequencing datasets to identify sequence variation in genes and enhancer elements in congenic and backcross mouse models. We defined genetic variation in the widely used B6-CD45.2 and B6.SJL-CD45.1 congenic model, identifying substantial differences in SJL genetic content retained in B6.SJL-CD45.1 strains on the basis of the vendor source of the mice. Genes encoding PD-1, CD62L, Bcl-2, cathepsin E, and Cxcr4 were within SJL genetic content in at least one vendor source of B6.SJL-CD45.1 mice. SJL genetic content affected enhancer elements, gene regulation, protein expression, and amino acid content in CD4+ T helper 1 cells, and mice infected with influenza showed reduced expression of Cxcr4 on B6.SJL-CD45.1 T follicular helper cells. These findings provide information on experimental variables and aid in creating approaches that account for genetic variables.
Collapse
Affiliation(s)
- Danielle A Chisolm
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Wayne Cheng
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Shelby A Colburn
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Aaron Silva-Sanchez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Selene Meza-Perez
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Troy D Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amy S Weinmann
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
11
|
Jafri S, Moore SD, Morrell NW, Ormiston ML. A sex-specific reconstitution bias in the competitive CD45.1/CD45.2 congenic bone marrow transplant model. Sci Rep 2017; 7:3495. [PMID: 28615666 PMCID: PMC5471196 DOI: 10.1038/s41598-017-03784-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/03/2017] [Indexed: 01/02/2023] Open
Abstract
Allelic variants of the pan-haematopoietic cell marker CD45, identified as CD45.1 and CD45.2, have been established as a marker system to track haematopoietic cells following congenic mouse bone marrow transplants. Despite the frequent use of this model for studying the impact of genetic modifications on relative differentiation potential, it is now evident that a bias exists in CD45.1 versus CD45.2 cell reconstitution. While this bias has been demonstrated by reduced reconstitution potential in B cells of CD45.1 origin, differences in the development of other lymphocytes, as well as the impact of sex on this bias, remain uncertain. We performed bone marrow transplants with wild-type CD45.1 and CD45.2 donor cells, and characterised haematopoietic cell reconstitution in dual-expressing CD45.1/2 host mice. We report an increase in CD45.2 reconstitution in the bone marrow that persists in the spleen, thymus and blood. Through the use of CD45.1/2 hosts, we demonstrate the intrinsic bias towards CD45.2 reconstitution is independent of an immunogenic response to the CD45.1 epitope. Furthermore, we identify a sex-specific difference in reconstitution efficiencies, with female mice exhibiting a greater bias towards CD45.2 reconstitution than males. This work sheds new light on the limitations of the CD45.1/CD45.2 congenic system for tracking lymphocyte development.
Collapse
Affiliation(s)
- Salema Jafri
- University of Cambridge, Department of Medicine, Cambridge, CB2 0QQ, United Kingdom
| | - Stephen D Moore
- University of Cambridge, Department of Medicine, Cambridge, CB2 0QQ, United Kingdom
| | - Nicholas W Morrell
- University of Cambridge, Department of Medicine, Cambridge, CB2 0QQ, United Kingdom
| | - Mark L Ormiston
- Queen's University, Department of Biomedical and Molecular Sciences, Kingston, K7L 3N6, Canada.
| |
Collapse
|
12
|
Stoeckle C, Geering B, Yousefi S, Rožman S, Andina N, Benarafa C, Simon HU. RhoH is a negative regulator of eosinophilopoiesis. Cell Death Differ 2016; 23:1961-1972. [PMID: 27740624 DOI: 10.1038/cdd.2016.73] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 06/16/2016] [Accepted: 06/27/2016] [Indexed: 02/07/2023] Open
Abstract
Eosinophils are frequently elevated in pathological conditions and can cause tissue damage and disease exacerbation. The number of eosinophils in the blood is largely regulated by factors controlling their production in the bone marrow. While several exogenous factors, such as interleukin-5, have been described to promote eosinophil differentiation, comparatively little is known about eosinophil-intrinsic factors that control their de novo generation. Here, we report that the small atypical GTPase RhoH is induced during human eosinophil differentiation, highly expressed in mature blood eosinophils and further upregulated in patients suffering from a hypereosinophilic syndrome. Overexpression of RhoH increases, in a Rho-associated protein kinase-dependent manner, the expression of GATA-2, a transcription factor involved in regulating eosinophil differentiation. In RhoH-/- mice, we observed reduced GATA-2 expression as well as accelerated eosinophil differentiation both in vitro and in vivo. Conversely, RhoH overexpression in bone marrow progenitors reduces eosinophil development in mixed bone marrow chimeras. These results highlight a novel negative regulatory role for RhoH in eosinophil differentiation, most likely in consequence of altered GATA-2 levels.
Collapse
Affiliation(s)
| | - Barbara Geering
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Saša Rožman
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Nicola Andina
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Charaf Benarafa
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Lu L, Dong J, Li D, Zhang J, Fan S. 3,3'-diindolylmethane mitigates total body irradiation-induced hematopoietic injury in mice. Free Radic Biol Med 2016; 99:463-471. [PMID: 27609226 DOI: 10.1016/j.freeradbiomed.2016.09.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/26/2016] [Accepted: 09/04/2016] [Indexed: 01/28/2023]
Abstract
We have reported that hematopoietic system injury induced by total body irradiation (TBI) leads to generation of intracellular reactive oxygen species (ROS) and DNA damage, which are ameliorated by antioxidant agents. In the present study, we reported that administration of DIM, a potent antioxidant agent, not only protected mice against TBI-induced lethality, also ameliorated TBI-induced hematopoietic injury. The latter effect was probably attributable to DIM's inhibition of TBI-induced increases in ROS production in hematopoietic stem cells (HSCs) and the phosphorylation of histone H2AX (γ-H2AX). In particular, DIM led to significant improvements in bone marrow (BM) HSC frequency, hematopoietic progenitor cell (HPC) clonogenic function, and multilineage engraftment after transplantation. A downregulation of NADPH oxidase 4 (NOX4) and an upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression were observed following DIM treatment. Notably, the anti-apoptotic potential of DIM was correlated with increased expression of the anti-apoptotic protein Bcl-2 and decreased expression of the pro-apoptotic protein Bax. These findings suggest that DIM attenuates TBI-induced hematopoietic injury through the inhibition of both oxidative stress in HSCs and hematopoietic cell apoptosis. Furthermore, we demonstrated that DIM protected BM hematopoietic cells against ionizing radiation and led to increased clonogenicity in vitro. Therefore, DIM has the potential to be used as an effective radioprotectant to ameliorate TBI-induced hematopoietic injury.
Collapse
Affiliation(s)
- Lu Lu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Junling Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
14
|
Murakami JL, Xu B, Franco CB, Hu X, Galli SJ, Weissman IL, Chen CC. Evidence that β7 Integrin Regulates Hematopoietic Stem Cell Homing and Engraftment Through Interaction with MAdCAM-1. Stem Cells Dev 2015; 25:18-26. [PMID: 26422691 DOI: 10.1089/scd.2014.0551] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
α4β7 integrin is a cell adhesion receptor that is crucial for the migration of hematopoietic progenitors and mature effector cells in the periphery, but its role in adult hematopoiesis is controversial. We identified a subset of hematopoietic stem cells (HSCs) in the bone marrow (BM) that expressed β7 integrin. These β7(+) HSCs were capable of multilineage, long-term reconstitution and had an inherent competitive advantage over β7(-) HSCs. On the other hand, HSCs that lacked β7 integrin (β7KO) had reduced engraftment potential. Interestingly, quantitative RT-PCR and flow cytometry revealed that β7KO HSCs expressed lower levels of the chemokine receptor CXCR4. Accordingly, β7KO HSCs exhibited impaired migration abilities in vitro and BM homing capabilities in vivo. Lethal irradiation induced expression of the α4β7 integrin ligand-mucosal addressin cell adhesion molecule-1 (MAdCAM-1) on BM endothelial cells. Moreover, blocking MAdCAM-1 reduced the homing of HSCs and impaired the survival of recipient mice. Altogether, these data indicate that β7 integrin, when expressed by HSCs, interacted with its endothelial ligand MAdCAM-1 in the BM microenvironment, thereby promoting HSC homing and engraftment.
Collapse
Affiliation(s)
- Jodi L Murakami
- 1 Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute of City of Hope , Duarte, California.,2 City of Hope Irell & Manella Graduate School of Biological Sciences , Duarte, California.,3 Gehr Family Center for Leukemia Research at City of Hope , Duarte, California
| | - Baohui Xu
- 4 Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Christopher B Franco
- 5 Department of Pathology, Stanford University School of Medicine , Stanford, California.,6 Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Xingbin Hu
- 1 Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute of City of Hope , Duarte, California.,7 Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University , Xi'an, People's Republic of China
| | - Stephen J Galli
- 5 Department of Pathology, Stanford University School of Medicine , Stanford, California.,8 Department of Microbiology and Immunology, Stanford University School of Medicine , Stanford, California
| | - Irving L Weissman
- 5 Department of Pathology, Stanford University School of Medicine , Stanford, California.,6 Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine , Stanford, California
| | - Ching-Cheng Chen
- 1 Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute of City of Hope , Duarte, California.,2 City of Hope Irell & Manella Graduate School of Biological Sciences , Duarte, California.,3 Gehr Family Center for Leukemia Research at City of Hope , Duarte, California
| |
Collapse
|
15
|
Chudziak D, Spohn G, Karpova D, Dauber K, Wiercinska E, Miettinen JA, Papayannopoulou T, Bönig H. Functional consequences of perturbed CXCL12 signal processing: analyses of immature hematopoiesis in GRK6-deficient mice. Stem Cells Dev 2014; 24:737-46. [PMID: 25316534 DOI: 10.1089/scd.2014.0284] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) reside in bone marrow (BM) in an environment rich in CXCL12, the ligand for CXCR4, which is constitutively expressed on all immature hematopoietic cells in BM. This ligand-receptor pair critically controls HSPC retention and (relative) quiescence in BM. Interestingly, in a chemokine-abundant environment, CXCR4 surface expression and CXCL12 sensitivity of BM-residing HSPCs are continuously maintained. The mechanisms underlying this peculiar pattern of G-protein signal integration by BM-HSPCs are unknown. G-protein receptor kinases (GRKs) control receptor function by phosphorylating the intracellular domains upon ligand-induced activation, which results in receptor internalization and transient refractoriness. Using, therefore, a GRK6-deficient (GRK6(-/-)) mouse, we sought to address how perturbed ligand-induced CXCR4 (in)activation affects HSPC behavior in vitro and in vivo. In vitro, GRK6(-/-) HSPCs were characterized by hyper-responsiveness to CXCL12, as expected. In vivo, GRK6(-/-) immature hematopoiesis was characterized by a marked expansion of immature hematopoiesis in spleens and a modest repopulation defect in serial competitive transplantation. Enforced mobilization with granulocyte colony-stimulating factor (G-CSF) and AMD3100 was normal, as was hematopoietic regeneration after noncompetitive transplantation or pharmacological myelosuppression. These observations illustrate that GRK-mediated restriction of CXCR4 signal input after ligand engagement is largely dispensable for BM-resident HSPCs, which may explain how continuous CXCL12 responsiveness of BM-HSPCs can be maintained.
Collapse
Affiliation(s)
- Doreen Chudziak
- 1 German Red Cross Blood Service Baden-Württemberg-Hesse , Frankfurt, Germany
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Comparative sensitivity analyses of quantitative polymerase chain reaction and flow cytometry in detecting cellular microchimerism in murine tissues. J Immunol Methods 2014; 406:74-82. [PMID: 24657636 DOI: 10.1016/j.jim.2014.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 01/30/2014] [Accepted: 03/11/2014] [Indexed: 11/23/2022]
Abstract
Cellular microchimerism is defined as the presence of small populations of cells from one individual in another genetically distinct individual. The pivotal role of cellular microchimerism in a variety of immune settings is increasingly recognized, e.g. in context of pregnancy, transplantation and cancer. However, the detection of chimeric cells is overshadowed by technical limitations. This study aimed to overcome these limitations by testing the sensitivity and detection limit of a molecular biology approach (quantitative polymerase chain reaction, qPCR) and a cellular approach (flow cytometry) in order to identify experimentally induced cellular microchimerism in mice. Leukocytes isolated from lymph nodes or spleens of transgenic enhanced green fluorescent protein (eGFP) and CD45.1 mice respectively were used as targets to be detected as microchimeric cells among wild type (wt) or haploidentical cells. The detection limit of microchimeric cells by flow cytometry was 0.05% or lower for the respective eGFP(+) or CD45.1(+) cell subsets, which equals 48 cells or fewer per 1×10(5) wt cells. The detection limit of CD45.1(+) and CD45.2(+) cells among haploidentical CD45.1(+)2(+) cells by flow cytometry was 48 cells (0.05%) and 198 cells (0.2%), respectively. Using qPCR, a detection limit of 198 eGFP(+) cells per 1×10(5) wt cells, respective 0.2%, could be achieved. We here introduce two technical approaches to reliably detect low number of chimeric cells at a low detection limit and high sensitivity in transgenic mouse systems.
Collapse
|