1
|
Zeng Z, Mao H, Lei Q, He Y. IL-7 in autoimmune diseases: mechanisms and therapeutic potential. Front Immunol 2025; 16:1545760. [PMID: 40313966 PMCID: PMC12043607 DOI: 10.3389/fimmu.2025.1545760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/14/2025] [Indexed: 05/03/2025] Open
Abstract
Interleukin-7 (IL-7) is a pleiotropic cytokine that plays a crucial role in the development, homeostasis, and function of the immune system. Growing evidence has demonstrated that IL-7 is involved in the pathogenesis of various autoimmune diseases including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D) and multiple sclerosis (MS). This review aims to summarize the current understanding of the role of IL-7 in autoimmune diseases, focusing on its mechanisms of action, implications for disease progression, and potential therapeutic applications. Produced by stromal cells, IL-7 binds to IL-7 receptor (IL-7R) on diverse immune cells. It is crucial for T cell development, survival, and proliferation. In autoimmune diseases, it activates and expands autoreactive T cells and influences B cell function, potentially leading to autoantibody production. The review further delves into the role of IL-7 in different autoimmune diseases. In RA, elevated IL-7/IL-7R promotes memory T cell survival, cytokine production, and influences B cells and monocytes to contribute to inflammation and joint damage. In SLE, elevated soluble form of IL-7R is associated with disease activity, promoting the survival of autoreactive T cells and enhancing the production of pro-inflammatory cytokines. In MS, genetic variations in the IL-7R gene are linked to disease susceptibility, and IL-7 impacts the survival and differentiation of T cell subsets involved in multiple sclerosis pathogenesis. For T1D, IL-7 affects the function of immune cells that attack pancreatic β cells. Given its central role in autoimmune processes, targeting the IL-7/IL-7R axis holds great therapeutic potential. By modulating IL-7 signaling, it may be possible to restore immune tolerance, reduce the activation of autoreactive immune cells, and alleviate disease symptoms. Understanding the complex mechanisms of IL-7 in autoimmune diseases is essential for the development of effective and targeted therapies.
Collapse
Affiliation(s)
| | | | | | - Yuanmin He
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Chen H, Zhou X, Liu T, Liu J, Wu D, Xu X, Ma S, Qiang G, Chen J, Cao Y, Fu W, Yang J. Postprandial parasympathetic signals promote lung type 2 immunity. Neuron 2025; 113:670-683.e7. [PMID: 39837323 DOI: 10.1016/j.neuron.2024.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 11/03/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025]
Abstract
Lung type 2 immunity protects against pathogenic infection, but its dysregulation causes asthma. Although it has long been observed that symptoms of asthmatic patients often become exaggerated following food intake, the pathophysiological mechanism underlying this postprandial phenomenon is incompletely understood. Here, we report that lung type 2 immunity in mice is enhanced after feeding, which correlates with parasympathetic activation. Also, local parasympathetic innervations exhibit spatial engagement with such immune responses mediated by group 2 innate lymphoid cells (ILC2s). Pharmacologic or surgical blockage of parasympathetic signals diminishes lung type 2 immunity. Conversely, chemogenetic manipulation of parasympathetic inputs and their upstream neurocircuit is sufficient to modulate those immune responses. We then show that the cholinergic receptor muscarinic 4 (Chrm4) for the parasympathetic neurotransmitter acetylcholine is expressed in mouse or human lung ILC2s, and the Chrm4 deletion mitigates ILC2-mediated lung inflammation. These results have revealed a critical neuroimmune function of the gut-brain-lung reflex.
Collapse
Affiliation(s)
- Hongjie Chen
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Xin Zhou
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; Department of General Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Tingting Liu
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Jiaqi Liu
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China
| | - Di Wu
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Xia Xu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shanwu Ma
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Guangliang Qiang
- Department of Thoracic Surgery, Peking University Third Hospital, Beijing 100191, China
| | - Jian Chen
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Ying Cao
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Wei Fu
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; Department of General Surgery, Peking University Third Hospital, Beijing 100191, China.
| | - Jing Yang
- PTN Graduate Program, Peking University Third Hospital Cancer Center, Center for Life Sciences, IDG/McGovern Institute for Brain Research, State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing 100871, China; Peking Union Medical College Hospital, Beijing 100730, China.
| |
Collapse
|
3
|
Ariail E, Biggs B, O'Flanagan R, Schneck JP. IL-7 Immunotherapies: Current Applications and Engineering Opportunities. Immunol Invest 2025:1-19. [PMID: 39981682 DOI: 10.1080/08820139.2025.2464055] [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] [Indexed: 02/22/2025]
Abstract
BACKGROUND IL-7 is a cytokine that plays a critical role in the development and proliferation of many different immune cells. IL-7 is notably important for the proper development and activity of T cells and B cells. Additionally, the cytokine plays a role in the function of natural killer cells and dendritic cells. Because of this innate biological activity, IL-7 has gained traction as a potential immunotherapy for multiple applications. METHODS We conducted a comprehensive literature review to explore the physiological role of IL-7 and current applications harnessing the biology of IL-7 as a therapeutic. We also investigated the ways in which IL-7 is being engineered to enhance its therapeutic potential. RESULTS Notably, IL-7 has demonstrated efficacy in adoptive cell therapy models and as a vaccine adjuvant. The cytokine has also been used as a treatment for sepsis and other chronic infections. To further enhance its therapeutic efficacy, IL-7 has been engineered by fusing the cytokine to antibody fragments or other bioactive or targeting molecules. These engineered IL-7 therapeutics seek to improve the cytokine's pharmacokinetic and immunological properties and reduce off-target effects. CONCLUSION IL-7 immunotherapies largely remain at the preclinical stage, but there is growing interest in IL-7's many therapeutic applications and increasing opportunities to further engineer the molecule for future clinical translation.
Collapse
Affiliation(s)
- Emily Ariail
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Benjamin Biggs
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rowan O'Flanagan
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan P Schneck
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Wang X, Fu M, Wang W, Shu S, Zhang N, Zhao R, Chen X, Hua X, Wang X, Feng W, Wang X, Song J. Single-cell analysis reveals the loss of FABP4-positive proliferating valvular endothelial cells relates to functional mitral regurgitation. BMC Med 2024; 22:595. [PMID: 39707349 DOI: 10.1186/s12916-024-03791-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 11/20/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Functional mitral regurgitation (MR) is a common form of mitral valve dysfunction that often persists even after surgical intervention, requiring reoperation in some cases. To advance our understanding of the pathogenesis of functional MR, it is crucial to characterize the cellular composition of the mitral valve leaflet and identify molecular changes in each cell subtype within the mitral valves of MR patients. Therefore, we aimed to comprehensively examine the cellular and molecular components of mitral valves in patients with MR. METHODS We conducted a single-cell RNA sequencing (scRNA-seq) analysis of mitral valve leaflets extracted from six patients who underwent heart transplantation. The cohort comprised three individuals with moderate-to-severe functional MR (MR group) and three non-diseased controls (NC group). Bioinformatics was applied to identify cell types, delineate cell functions, and explore cellular developmental trajectories and interactions. Key findings from the scRNA-seq analysis were validated using pathological staining to visualize key markers in the mitral valve leaflets. Additionally, in vitro experiments with human primary valvular endothelial cells were conducted to further support our results. RESULTS Our study revealed that valve interstitial cells are critical for adaptive valve remodelling, as they secrete extracellular matrix proteins and promote fibrosis. We discovered an abnormal decrease in a subpopulation of FABP4 (fatty acid binding protein 4)-positive proliferating valvular endothelial cells. The trajectory analysis identifies this subcluster as the origin of VECs. Immunohistochemistry on the expanded cohort showed a reduction of FABP4-positive VECs in patients with functional MR. Intervention experiments with primary cells indicated that FABP4 promotes proliferation and migration in mitral valve VECs and enhances TGFβ-induced differentiation. CONCLUSIONS Our study presented a comprehensive assessment of the mitral valve cellular landscape of patients with MR and sheds light on the molecular changes occurring in human mitral valves during functional MR. We found a notable reduction in the proliferating endothelial cell subpopulation of valve leaflets, and FABP4 was identified as one of their markers. Therefore, FABP4 positive VECs served as proliferating endothelial cells relates to functional mitral regurgitation. These VECs exhibited high proliferative and differentiative properties. Their reduction was associated with the occurrence of functional MR.
Collapse
Affiliation(s)
- Xiaohu Wang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
| | - Mengxia Fu
- Galactophore Department, Galactophore Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Weiteng Wang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
| | - Songren Shu
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Zhang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruojin Zhao
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
| | - Xiao Chen
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiumeng Hua
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Feng
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianqiang Wang
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China.
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jiangping Song
- Present Address: State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Beijing, Xi Cheng District, 100037, China.
- The Cardiomyopathy Research Group, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Cardiovascular Surgery, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, China.
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Cardiac Surgery, Fuwai Yunnan Hospital, Chinese Academy of Medical Sciences, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
5
|
Manso BA, Medina P, Smith-Berdan S, Rodriguez Y Baena A, Bachinsky E, Mok L, Deguzman A, Avila SB, Chattopadhyaya S, Rommel MGE, Jönsson VD, Forsberg EC. A rare HSC-derived megakaryocyte progenitor accumulates via enhanced survival and contributes to exacerbated thrombopoiesis upon aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621964. [PMID: 39574585 PMCID: PMC11580903 DOI: 10.1101/2024.11.04.621964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
Distinct routes of cellular production from hematopoietic stem cells (HSCs) have defined our current view of hematopoiesis. Recently, we challenged classical views of platelet generation, demonstrating that megakaryocyte progenitors (MkPs), and ultimately platelets, can be specified via an alternate and additive route of HSC-direct specification specifically during aging. This "shortcut" pathway generates hyperactive platelets likely to contribute to age-related platelet-mediated morbidities. Here, we used single-cell RNA/CITEseq to demonstrate that these age-unique, non-canonical (nc)MkPs can be prospectively defined and experimentally isolated from wild type mice. Surprisingly, this revealed that a rare population of ncMkPs also exist in young mice. Young and aged ncMkPs are functionally distinct from their canonical (c)MkP counterparts, with aged ncMkPs paradoxically and uniquely exhibiting enhanced survival and platelet generation capacity. We further demonstrate that aged HSCs generate significantly more ncMkPs than their younger counterparts, yet this is accomplished without strict clonal restriction. Together, these findings reveal significant phenotypic, functional, and aging-dependent heterogeneity among the MkP pool and uncover unique features of megakaryopoiesis throughout life, potentially offering cellular and molecular targets for mitigation of age-related adverse thrombotic events.
Collapse
|
6
|
Goates M, Shrestha A, Thapa S, Bettini M, Barrios R, Shivanna B. Blocking IL-17a Signaling Decreases Lung Inflammation and Improves Alveolarization in Experimental Bronchopulmonary Dysplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2023-2035. [PMID: 39117111 DOI: 10.1016/j.ajpath.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/13/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is the most common chronic lung disease of preterm infants that is associated with life-long morbidities. Inflammatory insults contribute to BPD pathogenesis. Although the proinflammatory cytokine, IL-17a, plays a role in various neonatal inflammatory disorders, its role in BPD pathogenesis is unclear. To test the hypothesis that blocking IL-17a signaling decreases lipopolysaccharide (LPS)-mediated experimental BPD in neonatal mice, wild-type mice were injected intraperitoneally with phosphate-buffered saline or LPS during the saccular lung developmental phase. Pulmonary IL-17a expression was determined by enzyme-linked immunosorbent assay and by flow cytometry. LPS-injected mice had higher pulmonary IL-17a protein levels and IL-17a+ and IL-22+ cells. γδ T cells, followed by non-T lymphoid cells, were the primary producers of IL-17a. Wild-type mice were then injected intraperitoneally with isotype antibody (Ab) or IL-17a Ab, while they were treated with phosphate-buffered saline or LPS, followed by quantification of lung inflammatory markers, alveolarization, vascularization, cell proliferation, and apoptosis. LPS-mediated alveolar simplification, apoptosis, and cell proliferation inhibition were significantly greater in mice treated with isotype Ab than in those treated with IL-17a Ab. Furthermore, STAT1 activation and IL-6 levels were significantly greater in LPS-exposed mice treated with isotype Ab than in those treated with IL-17a Ab. The study results indicate that blocking IL-17a signaling decreases LPS-mediated experimental BPD.
Collapse
Affiliation(s)
- Meagan Goates
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amrit Shrestha
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Matthew Bettini
- Department of Microbiology and Immunology, University of Utah, Salt Lake City, Utah
| | - Roberto Barrios
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
7
|
Yazdani R, Askari M, Ahmadi AM, Azizi G, Ciric B, Boehm A, Zhang GX, Rostami A. Mouse monocytes express CD127 by immune cells, not LPS. Front Immunol 2024; 15:1356004. [PMID: 39328413 PMCID: PMC11424446 DOI: 10.3389/fimmu.2024.1356004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The essential role of interleukin 7 (IL-7) signaling via its receptor (IL-7Rα; CD127) in T cell development and function has been well documented. However, CD127 expression and function in myeloid cells, including monocytes, are less clear, especially in mice. In the present study we report an inducible CD127 expression in mouse monocytes/macrophages. This induction is dependent on the presence of other immune cells, highlighting that regulation of CD127 expression on monocytes differs in mice and humans. We demonstrate that CD127 is functional, as IL-7 downregulated its expression. We also saw decreased CD127 expression during inflammation in vivo. Overall, upregulation of CD127 expression in vitro and its downregulation in vivo confirm that CD127 is an inducible marker on mouse monocyte/macrophage cells, in contrast to findings recently published by others. Characterizing the role of CD127 signaling in myeloid cells in inflammatory disorders would be worthwhile in future study.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University,
Philadelphia, PA, United States
| |
Collapse
|
8
|
de Matos Rodrigues J, Lokhande L, Olsson LM, Hassan M, Johansson A, Janská A, Kumar D, Schmidt L, Nikkarinen A, Hollander P, Glimelius I, Porwit A, Gerdtsson AS, Jerkeman M, Ek S. CD163+ macrophages in mantle cell lymphoma induce activation of prosurvival pathways and immune suppression. Blood Adv 2024; 8:4370-4385. [PMID: 38959399 PMCID: PMC11375268 DOI: 10.1182/bloodadvances.2023012039] [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: 10/26/2023] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
ABSTRACT Mantle cell lymphoma (MCL) is dependent on a supportive tumor immune microenvironment (TIME) in which infiltration of CD163+ macrophages has a negative prognostic impact. This study explores how abundance and spatial localization of CD163+ cells are associated with the biology of MCL, using spatial multiomic investigations of tumor and infiltrating CD163+ and CD3+ cells. A total of 63 proteins were measured using GeoMx digital spatial profiling in tissue microarrays from 100 diagnostic MCL tissues. Regions of interest were selected in tumor-rich and tumor-sparse tissue regions. Molecular profiling of CD163+ macrophages, CD20+ MCL cells, and CD3+ T-cells was performed. To validate protein profiles, 1811 messenger RNAs were measured in CD20+ cells and 2 subsets of T cells. Image analysis was used to extract the phenotype and position of each targeted cell, thereby allowing the exploration of cell frequencies and cellular neighborhoods. Proteomic investigations revealed that CD163+ cells modulate their immune profile depending on their localization and that the immune inhibitory molecules, V-domain immunoglobulin suppressor of T-cell activation and B7 homolog 3, have higher expression in tumor-sparse than in tumor-rich tissue regions and that targeting should be explored. We showed that MCL tissues with more abundant infiltration of CD163+ cells have a higher proteomic and transcriptional expression of key components of the MAPK pathway. Thus, the MAPK pathway may be a feasible therapeutic target in patients with MCL with CD163+ cell infiltration. We further showed the independent and combined prognostic values of CD11c and CD163 beyond established risk factors.
Collapse
Affiliation(s)
| | | | - Lina M. Olsson
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - May Hassan
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | - Anna Janská
- Department of Immunotechnology, Lund University, Lund, Sweden
| | | | - Lina Schmidt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Anna Nikkarinen
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | - Peter Hollander
- Department of Immunology, Genetics and Pathology, Clinical and Experimental Pathology, Uppsala University, Uppsala, Sweden
| | - Ingrid Glimelius
- Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, Uppsala, Sweden
| | - Anna Porwit
- Division of Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | | | - Mats Jerkeman
- Division of Oncology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Sara Ek
- Department of Immunotechnology, Lund University, Lund, Sweden
| |
Collapse
|
9
|
Gu W, Eke C, Gonzalez Santiago E, Olaloye O, Konnikova L. Single-cell atlas of the small intestine throughout the human lifespan demonstrates unique features of fetal immune cells. Mucosal Immunol 2024; 17:599-617. [PMID: 38555026 PMCID: PMC11384551 DOI: 10.1016/j.mucimm.2024.03.011] [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: 11/02/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024]
Abstract
Proper development of mucosal immunity is critical for human health. Over the past decade, it has become evident that in humans, this process begins in utero. However, there are limited data on the unique features and functions of fetal mucosal immune cells. To address this gap, we integrated several single-cell ribonucleic acid sequencing datasets of the human small intestine (SI) to create an SI transcriptional atlas throughout the human life span, ranging from the first trimester to adulthood, with a focus on immune cells. Fetal SI displayed a complex immune landscape comprising innate and adaptive immune cells that exhibited distinct transcriptional programs from postnatal samples, especially compared with pediatric and adult samples. We identified shifts in myeloid populations across gestation and progression of memory T-cell states throughout the human lifespan. In particular, there was a marked shift of memory T cells from those with stem-like properties in the fetal samples to fully differentiated cells with a high expression of activation and effector function genes in adult samples, with neonatal samples containing both features. Finally, we demonstrate that the SI developmental atlas can be used to elucidate improper trajectories linked to mucosal diseases by implicating developmental abnormalities underlying necrotizing enterocolitis, a severe intestinal complication of prematurity. Collectively, our data provide valuable resources and important insights into intestinal immunity that will facilitate regenerative medicine and disease understanding.
Collapse
Affiliation(s)
- Weihong Gu
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Chino Eke
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | | | - Oluwabunmi Olaloye
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA; Department of Obstetrics, Gynecology and Reproductive Science, Yale University School of Medicine, New Haven, CT, USA; Program in Translational Biomedicine, Yale University School of Medicine, New Haven, CT, USA; Program in Human Translational Immunology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
10
|
Poscablo DM, Worthington AK, Smith-Berdan S, Rommel MGE, Manso BA, Adili R, Mok L, Reggiardo RE, Cool T, Mogharrab R, Myers J, Dahmen S, Medina P, Beaudin AE, Boyer SW, Holinstat M, Jonsson VD, Forsberg EC. An age-progressive platelet differentiation path from hematopoietic stem cells causes exacerbated thrombosis. Cell 2024; 187:3090-3107.e21. [PMID: 38749423 PMCID: PMC12047039 DOI: 10.1016/j.cell.2024.04.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 02/05/2024] [Accepted: 04/16/2024] [Indexed: 06/09/2024]
Abstract
Platelet dysregulation is drastically increased with advanced age and contributes to making cardiovascular disorders the leading cause of death of elderly humans. Here, we reveal a direct differentiation pathway from hematopoietic stem cells into platelets that is progressively propagated upon aging. Remarkably, the aging-enriched platelet path is decoupled from all other hematopoietic lineages, including erythropoiesis, and operates as an additional layer in parallel with canonical platelet production. This results in two molecularly and functionally distinct populations of megakaryocyte progenitors. The age-induced megakaryocyte progenitors have a profoundly enhanced capacity to engraft, expand, restore, and reconstitute platelets in situ and upon transplantation and produce an additional platelet population in old mice. The two pools of co-existing platelets cause age-related thrombocytosis and dramatically increased thrombosis in vivo. Strikingly, aging-enriched platelets are functionally hyper-reactive compared with the canonical platelet populations. These findings reveal stem cell-based aging as a mechanism for platelet dysregulation and age-induced thrombosis.
Collapse
Affiliation(s)
- Donna M Poscablo
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, 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 95064, USA; Program in Biomedical Science and Engineering, 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 95064, USA; Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Marcel G E Rommel
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Bryce A Manso
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lydia Mok
- Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Roman E Reggiardo
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Taylor Cool
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Raana Mogharrab
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jenna Myers
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Steven Dahmen
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Paloma Medina
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Anna E Beaudin
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Scott W Boyer
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Program in Biomedical Science and Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vanessa D Jonsson
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Applied Mathematics, Genomics Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.
| |
Collapse
|
11
|
Manso BA, Rodriguez y Baena A, Forsberg EC. From Hematopoietic Stem Cells to Platelets: Unifying Differentiation Pathways Identified by Lineage Tracing Mouse Models. Cells 2024; 13:704. [PMID: 38667319 PMCID: PMC11048769 DOI: 10.3390/cells13080704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Platelets are the terminal progeny of megakaryocytes, primarily produced in the bone marrow, and play critical roles in blood homeostasis, clotting, and wound healing. Traditionally, megakaryocytes and platelets are thought to arise from multipotent hematopoietic stem cells (HSCs) via multiple discrete progenitor populations with successive, lineage-restricting differentiation steps. However, this view has recently been challenged by studies suggesting that (1) some HSC clones are biased and/or restricted to the platelet lineage, (2) not all platelet generation follows the "canonical" megakaryocytic differentiation path of hematopoiesis, and (3) platelet output is the default program of steady-state hematopoiesis. Here, we specifically investigate the evidence that in vivo lineage tracing studies provide for the route(s) of platelet generation and investigate the involvement of various intermediate progenitor cell populations. We further identify the challenges that need to be overcome that are required to determine the presence, role, and kinetics of these possible alternate pathways.
Collapse
Affiliation(s)
- Bryce A. Manso
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Alessandra Rodriguez y Baena
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
- Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - E. Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
- Department of Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
12
|
Abramson J, Dobeš J, Lyu M, Sonnenberg GF. The emerging family of RORγt + antigen-presenting cells. Nat Rev Immunol 2024; 24:64-77. [PMID: 37479834 PMCID: PMC10844842 DOI: 10.1038/s41577-023-00906-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 07/23/2023]
Abstract
Antigen-presenting cells (APCs) are master regulators of the immune response by directly interacting with T cells to orchestrate distinct functional outcomes. Several types of professional APC exist, including conventional dendritic cells, B cells and macrophages, and numerous other cell types have non-classical roles in antigen presentation, such as thymic epithelial cells, endothelial cells and granulocytes. Accumulating evidence indicates the presence of a new family of APCs marked by the lineage-specifying transcription factor retinoic acid receptor-related orphan receptor-γt (RORγt) and demonstrates that these APCs have key roles in shaping immunity, inflammation and tolerance, particularly in the context of host-microorganism interactions. These RORγt+ APCs include subsets of group 3 innate lymphoid cells, extrathymic autoimmune regulator-expressing cells and, potentially, other emerging populations. Here, we summarize the major findings that led to the discovery of these RORγt+ APCs and their associated functions. We discuss discordance in recent reports and identify gaps in our knowledge in this burgeoning field, which has tremendous potential to advance our understanding of fundamental immune concepts.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Jan Dobeš
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Mengze Lyu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
13
|
Ghinea FS, Ionică MV, Liliac IM, Pătru S, Olaru DG, Popa-Wagner A. The Impact of Juvenile Microglia Transcriptomics on the Adult Brain Regeneration after Cerebral Ischemia. CURRENT HEALTH SCIENCES JOURNAL 2024; 50:133-150. [PMID: 38846476 PMCID: PMC11151955 DOI: 10.12865/chsj.50.01.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/22/2024] [Indexed: 06/09/2024]
Abstract
Microglial cells play a pivotal role in the brain's health and operation through all stages of life and in the face of illness. The contributions of microglia during the developmental phase of the brain markedly contrast with their contributions in the brain of adults after injury. Enhancing our understanding of the pathological mechanisms that involve microglial activity in brains as they age and in cerebrovascular conditions is crucial for informing the creation of novel therapeutic approaches. In this work we provide results on microglia transcriptomics in the juvenile vs injured adult brain and its impact on adult brain regeneration after cerebral ischemia. During fetal brain development, microglia cells are involved in gliogenesis, angiogenesis, axonal outgrowth, synaptogenesis, neurogenesis and synaptic reorganization by engulfing neuronal extensions. Within the mature, intact brain, microglial cells exhibit reduced movement of their processes in response to minimal neuronal activity, while they continuously monitor their surroundings and clear away cellular debris. Following a stroke in the adult brain, inflammation, neurodegeneration, or disruptions in neural equilibrium trigger alterations in both the genetic blueprint and the structure and roles of microglia, a state often described as "activated" microglia. Such genetic shifts include a notable increase in the pathways related to phagosomes, lysosomes, and the presentation of antigens, coupled with a rise in the expression of genes linked to cell surface receptors. We conclude that a comparison of microglia transcriptomic activity during brain development and post-stroke adult brain might provide us with new clues about how neurodegeneration occurs in the adult brain. This information could very useful to develop drugs to slow down or limit the post-stroke pathology and improve clinical outcome.
Collapse
Affiliation(s)
- Flavia Semida Ghinea
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Medicine Craiova, Romania
| | - Marius Viorel Ionică
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Medicine Craiova, Romania
| | | | - Simion Pătru
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Medicine Craiova, Romania
| | - Denisa Greta Olaru
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Medicine Craiova, Romania
| | - Aurel Popa-Wagner
- Experimental Research Center for Normal and Pathological Aging, University of Medicine and Medicine Craiova, Romania
| |
Collapse
|
14
|
Fransen LFH, Leonard MO. Induced pluripotent and CD34+ stem cell derived myeloid cells display differential responses to particle and dust mite exposure. Sci Rep 2023; 13:9375. [PMID: 37296179 PMCID: PMC10256772 DOI: 10.1038/s41598-023-36508-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/05/2023] [Indexed: 06/12/2023] Open
Abstract
Myeloid cells form an essential component of initial responses to environmental hazards and toxic exposures. The ability to model these responses in vitro is central to efforts tasked with identifying hazardous materials and understanding mechanisms of injury and disease. Induced pluripotent stem cell (iPSC) derived cells have been suggested as alternatives to more established primary cell testing systems for these purposes. iPSC derived macrophage and dendritic like cells were compared to CD34+ haematopoietic stem cell derived populations using transcriptomic analysis. Using single cell sequencing-based characterisation of iPSC derived myeloid cells, we identified transitional, mature and M2 like macrophages as well as dendritic like antigen presenting cells and fibrocytes. Direct transcriptomic comparisons between iPSC and CD34+ cell derived populations revealed higher expression of myeloid differentiation genes such as MNDA, CSF1R and CSF2RB in CD34+ cells, while iPSC populations had higher fibroblastic and proliferative markers. Exposure of differentiated macrophage populations to nanoparticle alone or in combination with dust mite, resulted in differential gene expression on combination only, with responses markedly absent in iPSC compared to CD34+ derived cells. The lack of responsiveness in iPSC derived cells may be attributable to lower levels of dust mite component receptors CD14, TLR4, CLEC7A and CD36. In summary, iPSC derived myeloid cells display typical characteristics of immune cells but may lack a fully mature phenotype to adequately respond to environmental exposures.
Collapse
Affiliation(s)
- Leonie F H Fransen
- Toxicology Department, Radiation, Chemical and Environmental Hazards Directorate, UK Health Security Agency, Chilton, Harwell Campus, Didcot, OX11 0RQ, UK
| | - Martin O Leonard
- Toxicology Department, Radiation, Chemical and Environmental Hazards Directorate, UK Health Security Agency, Chilton, Harwell Campus, Didcot, OX11 0RQ, UK.
| |
Collapse
|
15
|
Pinosanu LR, Capitanescu B, Glavan D, Godeanu S, Cadenas IF, Doeppner TR, Hermann DM, Balseanu AT, Bogdan C, Popa-Wagner A. Neuroglia Cells Transcriptomic in Brain Development, Aging and Neurodegenerative Diseases. Aging Dis 2023; 14:63-83. [PMID: 36818562 PMCID: PMC9937697 DOI: 10.14336/ad.2022.0621] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/21/2022] [Indexed: 11/18/2022] Open
Abstract
Glia cells are essential for brain functioning during development, aging and disease. However, the role of astroglia plays during brain development is quite different from the role played in the adult lesioned brain. Therefore, a deeper understanding of pathomechanisms underlying astroglia activity in the aging brain and cerebrovascular diseases is essential to guide the development of new therapeutic strategies. To this end, this review provides a comparison between the transcriptomic activity of astroglia cells during development, aging and neurodegenerative diseases, including cerebral ischemia. During fetal brain development, astrocytes and microglia often affect the same developmental processes such as neuro-/gliogenesis, angiogenesis, axonal outgrowth, synaptogenesis, and synaptic pruning. In the adult brain astrocytes are a critical player in the synapse remodeling by mediating synapse elimination while microglia activity has been associated with changes in synaptic plasticity and remove cell debris by constantly sensing the environment. However, in the lesioned brain astrocytes proliferate and play essential functions with regard to energy supply to the neurons, neurotransmission and buildup of a protective scar isolating the lesion site from the surroundings. Inflammation, neurodegeneration, or loss of brain homeostasis induce changes in microglia gene expression, morphology, and function, generally referred to as "primed" microglia. These changes in gene expression are characterized by an enrichment of phagosome, lysosome, and antigen presentation signaling pathways and is associated with an up-regulation of genes encoding cell surface receptors. In addition, primed microglia are characterized by upregulation of a network of genes in response to interferon gamma. Conclusion. A comparison of astroglia cells transcriptomic activity during brain development, aging and neurodegenerative disorders might provide us with new therapeutic strategies with which to protect the aging brain and improve clinical outcome.
Collapse
Affiliation(s)
- Leonard Radu Pinosanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Bogdan Capitanescu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Daniela Glavan
- Psychiatric clinic, University of Medicine and Pharmacy Craiova, Craiova, Romania.
| | - Sanziana Godeanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Israel Ferna´ndez Cadenas
- Stroke Pharmacogenomics and Genetics group, Sant Pau Hospital Institute of Research, Barcelona, Spain.
| | - Thorsten R. Doeppner
- Department of Neurology, University Hospital Giessen, Giessen, Germany.,University of Göttingen Medical School, Department of Neurology, Göttingen, Germany.
| | - Dirk M. Hermann
- Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.
| | - Adrian-Tudor Balseanu
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.
| | - Catalin Bogdan
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.,Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.,Correspondence should be addressed to: Dr. Aurel Popa-Wagner () and Dr. Catalin Bogdan (), University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Aurel Popa-Wagner
- Experimental Research Center for Normal and Pathological Aging (ARES), University of Medicine and Pharmacy of Craiova, Craiova, Romania.,Vascular Neurology, Dementia and Ageing Research, Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, Germany.,Correspondence should be addressed to: Dr. Aurel Popa-Wagner () and Dr. Catalin Bogdan (), University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147 Essen, Germany
| |
Collapse
|
16
|
Fernandes MB, Barata JT. IL-7 and IL-7R in health and disease: An update through COVID times. Adv Biol Regul 2023; 87:100940. [PMID: 36503870 DOI: 10.1016/j.jbior.2022.100940] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
The role of IL-7 and IL-7R for normal lymphoid development and an adequately functioning immune system has been recognized for long, with severe immune deficiency and lymphoid leukemia as extreme examples of the consequences of deregulation of the IL-7-IL-7R axis. In this review, we provide an update (focusing on the past couple of years) on IL-7 and IL-7R in health and disease. We highlight the findings on IL-7/IL-7R signaling mechanisms and the, sometimes controversial, impact of IL-7 and its receptor on leukocyte biology, COVID-19, acute lymphoblastic leukemia, and different solid tumors, as well as their relevance as therapeutic tools or targets.
Collapse
Affiliation(s)
- Marta B Fernandes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028, Lisbon, Portugal.
| |
Collapse
|
17
|
IL-7: Comprehensive review. Cytokine 2022; 160:156049. [DOI: 10.1016/j.cyto.2022.156049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 01/08/2023]
|
18
|
Brace N, Megson IL, Rossi AG, Doherty MK, Whitfield PD. SILAC-based quantitative proteomics to investigate the eicosanoid associated inflammatory response in activated macrophages. J Inflamm (Lond) 2022; 19:12. [PMID: 36050729 PMCID: PMC9438320 DOI: 10.1186/s12950-022-00309-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Macrophages play a central role in inflammation by phagocytosing invading pathogens, apoptotic cells and debris, as well as mediating repair of tissues damaged by trauma. In order to do this, these dynamic cells generate a variety of inflammatory mediators including eicosanoids such as prostaglandins, leukotrienes and hydroxyeicosatraenoic acids (HETEs) that are formed through the cyclooxygenase, lipoxygenase and cytochrome P450 pathways. The ability to examine the effects of eicosanoid production at the protein level is therefore critical to understanding the mechanisms associated with macrophage activation. RESULTS This study presents a stable isotope labelling with amino acids in cell culture (SILAC) -based proteomics strategy to quantify the changes in macrophage protein abundance following inflammatory stimulation with Kdo2-lipid A and ATP, with a focus on eicosanoid metabolism and regulation. Detailed gene ontology analysis, at the protein level, revealed several key pathways with a decrease in expression in response to macrophage activation, which included a promotion of macrophage polarisation and dynamic changes to energy requirements, transcription and translation. These findings suggest that, whilst there is evidence for the induction of a pro-inflammatory response in the form of prostaglandin secretion, there is also metabolic reprogramming along with a change in cell polarisation towards a reduced pro-inflammatory phenotype. CONCLUSIONS Advanced quantitative proteomics in conjunction with functional pathway network analysis is a useful tool to investigate the molecular pathways involved in inflammation.
Collapse
Affiliation(s)
- Nicole Brace
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Ian L Megson
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Mary K Doherty
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Phillip D Whitfield
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK.
- Present Address: Glasgow Polyomics, Garscube Campus, University of Glasgow, Glasgow, G61 1BD, UK.
| |
Collapse
|
19
|
Tang Y, Kwiatkowski DJ, Henske EP. Midkine expression by stem-like tumor cells drives persistence to mTOR inhibition and an immune-suppressive microenvironment. Nat Commun 2022; 13:5018. [PMID: 36028490 PMCID: PMC9418323 DOI: 10.1038/s41467-022-32673-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
mTORC1 is hyperactive in multiple cancer types1,2. Here, we performed integrative analysis of single cell transcriptomic profiling, paired T cell receptor (TCR) sequencing, and spatial transcriptomic profiling on Tuberous Sclerosis Complex (TSC) associated tumors with mTORC1 hyperactivity, and identified a stem-like tumor cell state (SLS) linked to T cell dysfunction via tumor-modulated immunosuppressive macrophages. Rapamycin and its derivatives (rapalogs) are the primary treatments for TSC tumors, and the stem-like tumor cells showed rapamycin resistance in vitro, reminiscent of the cytostatic effects of these drugs in patients. The pro-angiogenic factor midkine (MDK) was highly expressed by the SLS population, and associated with enrichment of endothelial cells in SLS-dominant samples. Inhibition of MDK showed synergistic benefit with rapamycin in reducing the growth of TSC cell lines in vitro and in vivo. In aggregate, this study suggests an autocrine rapamycin resistance mechanism and a paracrine tumor survival mechanism via immune suppression adopted by the stem-like state tumor cells with mTORC1 hyperactivity.
Collapse
Affiliation(s)
- Yan Tang
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David J Kwiatkowski
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Worthington AK, Cool T, Poscablo DM, Hussaini A, Beaudin AE, Forsberg EC. IL7Rα, but not Flk2, is required for hematopoietic stem cell reconstitution of tissue-resident lymphoid cells. Development 2022; 149:274067. [PMID: 35072209 PMCID: PMC8917444 DOI: 10.1242/dev.200139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022]
Abstract
Tissue-resident lymphoid cells (TLCs) span the spectrum of innate-to-adaptive immune function. Unlike traditional, circulating lymphocytes that are continuously generated from hematopoietic stem cells (HSCs), many TLCs are of fetal origin and poorly generated from adult HSCs. Here, we sought to further understand murine TLC development and the roles of Flk2 and IL7Rα, two cytokine receptors with known function in traditional lymphopoiesis. Using Flk2- and Il7r-Cre lineage tracing, we found that peritoneal B1a cells, splenic marginal zone B (MZB) cells, lung ILC2s and regulatory T cells (Tregs) were highly labeled. Despite high labeling, loss of Flk2 minimally affected the generation of these cells. In contrast, loss of IL7Rα, or combined deletion of Flk2 and IL7Rα, dramatically reduced the number of B1a cells, MZBs, ILC2s and Tregs, both in situ and upon transplantation, indicating an intrinsic and essential role for IL7Rα. Surprisingly, reciprocal transplants of wild-type HSCs showed that an IL7Rα−/− environment selectively impaired reconstitution of TLCs when compared with TLC numbers in situ. Taken together, our data defined Flk2- and IL7Rα-positive TLC differentiation paths, and revealed functional roles of Flk2 and IL7Rα in TLC establishment. Summary: Tissue-resident lymphoid cells develop via IL7Rα-positive progenitors and are repopulated by transplanted adult hematopoietic stem cells; however, such TLC lymphopoiesis cannot be fully rescued in IL7Rα−/− recipient mice.
Collapse
Affiliation(s)
- Atesh K Worthington
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Program in Biomedical Science and Engineering: Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Taylor Cool
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Program in Biomedical Science and Engineering: Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Donna M Poscablo
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Program in Biomedical Science and Engineering: Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Adeel Hussaini
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Anna E Beaudin
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California, Santa Cruz, Santa Cruz, CA 95064, USA.,Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
21
|
Wang X, Chang S, Wang T, Wu R, Huang Z, Sun J, Liu J, Yu Y, Mao Y. IL7R Is Correlated With Immune Cell Infiltration in the Tumor Microenvironment of Lung Adenocarcinoma. Front Pharmacol 2022; 13:857289. [PMID: 35264973 PMCID: PMC8899515 DOI: 10.3389/fphar.2022.857289] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/04/2022] [Indexed: 12/09/2022] Open
Abstract
Tumor microenvironment plays an important role in the development, progression, and prognosis of lung adenocarcinoma. Exploring new biomarkers based on the immune microenvironment of lung adenocarcinoma can effectively predict the prognosis and provide effective clinical treatment. In this study, we used the ESTIMATE algorithm to score the immune and stromal components in lung adenocarcinoma data downloaded from the TCGA database. The result showed that the immune/stromal score was associated with clinical features and prognosis of lung adenocarcinoma patients. Interleukin-7 receptor (IL7R) is an important prognostic biomarker identified by intersection analysis of protein-protein interaction networks and Cox regression survival analysis. According to TCGA and Oncomine database analysis, IL7R expression in adenocarcinoma tissues was significantly lower than that in normal lung tissues and was further verified in clinical tissue samples. Survival analysis showed IL7R was an independent prognostic factor of lung adenocarcinoma. IL7R expression was positively correlated with the overall survival and progression-free survival of lung adenocarcinoma patients and negatively correlated with tumor size. Our results suggest that IL7R inhibits tumor growth by regulating the proportion of immune infiltrating cells in the tumor immune microenvironment. IL7R could be a beneficial prognostic marker in patients with lung adenocarcinoma and has great potential in immune therapy.
Collapse
Affiliation(s)
- Xin Wang
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Shujian Chang
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Teng Wang
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Ruirong Wu
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Zebo Huang
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Junjie Sun
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jingjing Liu
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Yu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yong Mao
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
22
|
Li G, Zhang J, Liu Y, Cheng X, Sun K, Hong W, Sha K. Analyzing Prognostic Hub Genes in the Microenvironment of Cutaneous Melanoma by Computer Integrated Bioinformatics. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:4493347. [PMID: 35300397 PMCID: PMC8923759 DOI: 10.1155/2022/4493347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/22/2022] [Indexed: 11/18/2022]
Abstract
Cutaneous melanoma (CM) is attracting increasing attention due to high mortality. In response to this, we synthetically analyze the CM dataset from the TCGA database and explore microenvironment-related genes that effectively predict patient prognosis. Immune/stromal scores of cases are calculated using the ESTIMATE algorithm and are significantly associated with overall patient survival. Then, differentially expressed genes are identified by comparing the immune score and stromal score, also prognostic genes are subsequently screened. Functional analysis shows that these genes are enriched in different activities of immune system. Moreover, 19 prognosis-related hub genes are extracted from the protein-protein interaction network, of which four unreported genes (IL7R, FLT3, C1QC, and HLA-DRB5) are chosen for validation. A significant negative relationship is found between the expression levels of the 4 genes and pathological stages, notably T grade. Furthermore, the K-M plots and TIMER results show that these genes have favorable value for CM prognosis. In conclusion, these results give a novel insight into CM and identify IL7R, FLT3, C1QC, and HLA-DRB5 as crucial roles for the diagnosis and treatment of CM.
Collapse
Affiliation(s)
- Guangyao Li
- Affiliated Hospital of Jiujiang University, No. 57 Xunyang East Road, Jiujiang 332000, Jiangxi Province, China
| | - Jingye Zhang
- Affiliated Hospital of Jiujiang University, No. 57 Xunyang East Road, Jiujiang 332000, Jiangxi Province, China
| | - Yourao Liu
- Affiliated Hospital of Jiujiang University, No. 57 Xunyang East Road, Jiujiang 332000, Jiangxi Province, China
| | - Xiqing Cheng
- Affiliated Hospital of Jiujiang University, No. 57 Xunyang East Road, Jiujiang 332000, Jiangxi Province, China
| | - Kai Sun
- Affiliated Hospital of Jiujiang University, No. 57 Xunyang East Road, Jiujiang 332000, Jiangxi Province, China
| | - Wenjuan Hong
- Affiliated Hospital of Jiujiang University, No. 57 Xunyang East Road, Jiujiang 332000, Jiangxi Province, China
| | - Ke Sha
- First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
23
|
Rodriguez Y Baena A, Rajendiran S, Manso BA, Krietsch J, Boyer SW, Kirschmann J, Forsberg EC. New transgenic mouse models enabling pan-hematopoietic or selective hematopoietic stem cell depletion in vivo. Sci Rep 2022; 12:3156. [PMID: 35210475 PMCID: PMC8873235 DOI: 10.1038/s41598-022-07041-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/07/2022] [Indexed: 12/22/2022] Open
Abstract
Hematopoietic stem cell (HSC) multipotency and self-renewal are typically defined through serial transplantation experiments. Host conditioning is necessary for robust HSC engraftment, likely by reducing immune-mediated rejection and by clearing limited HSC niche space. Because irradiation of the recipient mouse is non-specific and broadly damaging, there is a need to develop alternative models to study HSC performance at steady-state and in the absence of radiation-induced stress. We have generated and characterized two new mouse models where either all hematopoietic cells or only HSCs can be specifically induced to die in vivo or in vitro. Hematopoietic-specific Vav1-mediated expression of a loxP-flanked diphtheria-toxin receptor (DTR) renders all hematopoietic cells sensitive to diphtheria toxin (DT) in “Vav-DTR” mice. Crossing these mice to Flk2-Cre mice results in “HSC-DTR” mice which exhibit HSC-selective DT sensitivity. We demonstrate robust, rapid, and highly selective cell ablation in these models. These new mouse models provide a platform to test whether HSCs are required for long-term hematopoiesis in vivo, for understanding the mechanisms regulating HSC engraftment, and interrogating in vivo hematopoietic differentiation pathways and mechanisms regulating hematopoietic homeostasis.
Collapse
Affiliation(s)
- Alessandra Rodriguez Y Baena
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA.,Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Smrithi Rajendiran
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA.,Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Bryce A Manso
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA.,Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Jana Krietsch
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA.,Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Scott W Boyer
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA.,Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA
| | - Jessica Kirschmann
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA. .,Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, 95064, USA.
| |
Collapse
|
24
|
Zhang B, Zhang Y, Xiong L, Li Y, Zhang Y, Zhao J, Jiang H, Li C, Liu Y, Liu X, Liu H, Ping YF, Zhang QC, Zhang Z, Bian XW, Zhao Y, Hu X. CD127 imprints functional heterogeneity to diversify monocyte responses in inflammatory diseases. J Exp Med 2022; 219:e20211191. [PMID: 35015026 PMCID: PMC8757045 DOI: 10.1084/jem.20211191] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/21/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory monocytes are key mediators of acute and chronic inflammation; yet, their functional diversity remains obscure. Single-cell transcriptome analyses of human inflammatory monocytes from COVID-19 and rheumatoid arthritis patients revealed a subset of cells positive for CD127, an IL-7 receptor subunit, and such positivity rendered otherwise inert monocytes responsive to IL-7. Active IL-7 signaling engaged epigenetically coupled, STAT5-coordinated transcriptional programs to restrain inflammatory gene expression, resulting in inverse correlation between CD127 expression and inflammatory phenotypes in a seemingly homogeneous monocyte population. In COVID-19 and rheumatoid arthritis, CD127 marked a subset of monocytes/macrophages that retained hypoinflammatory phenotypes within the highly inflammatory tissue environments. Furthermore, generation of an integrated expression atlas revealed unified features of human inflammatory monocytes across different diseases and different tissues, exemplified by those of the CD127high subset. Overall, we phenotypically and molecularly characterized CD127-imprinted functional heterogeneity of human inflammatory monocytes with direct relevance for inflammatory diseases.
Collapse
Affiliation(s)
- Bin Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Yuan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Lei Xiong
- Ministry of Education Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuzhe Li
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yunliang Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunological Diseases, Beijing, China
| | - Hui Jiang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunological Diseases, Beijing, China
| | - Can Li
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunological Diseases, Beijing, China
| | - Yunqi Liu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Xindong Liu
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haofei Liu
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi-Fang Ping
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiangfeng Cliff Zhang
- Ministry of Education Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, China
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiu-Wu Bian
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunological Diseases, Beijing, China
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Center for Human Disease Immuno-monitoring, Beijing Friendship Hospital, Beijing, China
| |
Collapse
|
25
|
Transcriptomic and Lipidomic Mapping of Macrophages in the Hub of Chronic Beta-Adrenergic-Stimulation Unravels Hypertrophy-, Proliferation-, and Lipid Metabolism-Related Genes as Novel Potential Markers of Early Hypertrophy or Heart Failure. Biomedicines 2022; 10:biomedicines10020221. [PMID: 35203431 PMCID: PMC8869621 DOI: 10.3390/biomedicines10020221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/13/2022] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
Sympathetic nervous system overdrive with chronic release of catecholamines is the most important neurohormonal mechanism activated to maintain cardiac output in response to heart stress. Beta-adrenergic signaling behaves first as a compensatory pathway improving cardiac contractility and maladaptive remodeling but becomes dysfunctional leading to pathological hypertrophy and heart failure (HF). Cardiac remodeling is a complex inflammatory syndrome where macrophages play a determinant role. This study aimed at characterizing the temporal transcriptomic evolution of cardiac macrophages in mice subjected to beta-adrenergic-stimulation using RNA sequencing. Owing to a comprehensive bibliographic analysis and complementary lipidomic experiments, this study deciphers typical gene profiles in early compensated hypertrophy (ECH) versus late dilated remodeling related to HF. We uncover cardiac hypertrophy- and proliferation-related transcription programs typical of ECH or HF macrophages and identify lipid metabolism-associated and Na+ or K+ channel-related genes as markers of ECH and HF macrophages, respectively. In addition, our results substantiate the key time-dependent role of inflammatory, metabolic, and functional gene regulation in macrophages during beta-adrenergic dependent remodeling. This study provides important and novel knowledge to better understand the prevalent key role of resident macrophages in response to chronically activated beta-adrenergic signaling, an effective diagnostic and therapeutic target in failing hearts.
Collapse
|
26
|
Cool T, Baena ARY, Forsberg EC. Clearing the Haze: How Does Nicotine Affect Hematopoiesis before and after Birth? Cancers (Basel) 2021; 14:184. [PMID: 35008347 PMCID: PMC8750289 DOI: 10.3390/cancers14010184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 11/24/2022] Open
Abstract
Hematopoiesis is a tightly regulated process orchestrated by cell-intrinsic and cell-extrinsic cues. Over the past several decades, much effort has been focused on understanding how these cues regulate hematopoietic stem cell (HSC) function. Many endogenous key regulators of hematopoiesis have been identified and extensively characterized. Less is known about the mechanisms of long-term effects of environmental toxic compounds on hematopoietic stem and progenitor cells (HSPCs) and their mature immune cell progeny. Research over the past several decades has demonstrated that tobacco products are extremely toxic and pose huge risks to human health by causing diseases like cancer, respiratory illnesses, strokes, and more. Recently, electronic cigarettes have been promoted as a safer alternative to traditional tobacco products and have become increasingly popular among younger generations. Nicotine, the highly toxic compound found in many traditional tobacco products, is also found in most electronic cigarettes, calling into question their purported "safety". Although it is known that nicotine is toxic, the pathophysiology of disease in exposed people remains under investigation. One plausible contributor to altered disease susceptibility is altered hematopoiesis and associated immune dysfunction. In this review, we focus on research that has addressed how HSCs and mature blood cells respond to nicotine, as well as identify remaining questions.
Collapse
Affiliation(s)
- Taylor Cool
- Program in Molecular, Cell, and Developmental Biology, Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, CA 95064, USA; (T.C.); (A.R.y.B.)
| | - Alessandra Rodriguez y Baena
- Program in Molecular, Cell, and Developmental Biology, Institute for the Biology of Stem Cells, University of California Santa Cruz, Santa Cruz, CA 95064, USA; (T.C.); (A.R.y.B.)
| | - E. Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
27
|
Rodriguez Y Baena A, Manso BA, Forsberg EC. CFU-S assay: a historical single-cell assay that offers modern insight into clonal hematopoiesis. Exp Hematol 2021; 104:1-8. [PMID: 34688837 DOI: 10.1016/j.exphem.2021.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 10/20/2022]
Abstract
Hematopoietic stem cells (HSCs) have been studied extensively since their initial functional description in 1961 when Dr. James Till and Dr. Ernest McCulloch developed the first in vivo clonal strategy, termed the spleen colony-forming unit (CFU-S) assay, to assess the functional capacity of bone marrow-derived hematopoietic progenitors at the single-cell level. Through transplantation of bone marrow cells and analysis of the resulting cellular nodules in the spleen, the CFU-S assay revealed both the self-renewal and clonal differentiation capacity of hematopoietic progenitors. Further development and use of this assay have identified highly proliferative, self-renewing, and differentiating HSCs that possess clonal, multilineage differentiation. The CFU-S strategy has also been adapted to interrogating single purified hematopoietic stem and progenitor cell populations, advancing our knowledge of the hematopoietic hierarchy. In this review, we explore the major discoveries made with the CFU-S assay, consider its modern use and recent improvements, and compare it with commonly used long-term transplantation assays to determine the continued value of the CFU-S assay for understanding HSC biology and hematopoiesis.
Collapse
Affiliation(s)
- Alessandra Rodriguez Y Baena
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA
| | - Bryce A Manso
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA.
| |
Collapse
|
28
|
Smith-Berdan S, Bercasio A, Kramer L, Petkus B, Hinck L, Forsberg EC. Acute and endothelial-specific Robo4 deletion affect hematopoietic stem cell trafficking independent of VCAM1. PLoS One 2021; 16:e0255606. [PMID: 34388149 PMCID: PMC8362960 DOI: 10.1371/journal.pone.0255606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/20/2021] [Indexed: 11/18/2022] Open
Abstract
Hematopoietic stem cell (HSC) trafficking is regulated by a number of complex mechanisms. Among them are the transmembrane protein Robo4 and the vascular cell adhesion molecule, VCAM1. Endothelial VCAM1 is a well-known regulator of hematopoietic cell trafficking, and our previous studies revealed that germline deletion of Robo4 led to impaired HSC trafficking, with an increase in vascular endothelial cell (VEC) numbers and downregulation of VCAM1 protein on sinusoidal VECs. Here, we utilized two Robo4 conditional deletion models in parallel with Robo4 germline knockout mice (R4KO) to evaluate the effects of acute and endothelial cell-specific Robo4 deletion on HSC trafficking. Strikingly similar to the R4KO, the acute deletion of Robo4 resulted in altered HSC distribution between the bone marrow and blood compartments, despite normal numbers of VECs and wild-type levels of VCAM1 cell surface protein on sinusoidal VECs. Additionally, consistent with the R4KO mice, acute loss of Robo4 in the host perturbed long-term engraftment of donor wild-type HSCs and improved HSC mobilization to the peripheral blood. These data demonstrate the significant role that endothelial Robo4 plays in directional HSC trafficking, independent of alterations in VEC numbers and VCAM1 expression.
Collapse
Affiliation(s)
- Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
- Department of Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - Alyssa Bercasio
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - Leah Kramer
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - Bryan Petkus
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - Lindsay Hinck
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
- Department of Molecular, Cell and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA, United States of America
| | - E. Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA, United States of America
- Department of Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA, United States of America
- * E-mail:
| |
Collapse
|
29
|
|
30
|
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: 23] [Impact Index Per Article: 5.8] [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
|
31
|
Data mining of immune-related prognostic genes in metastatic melanoma microenvironment. Biosci Rep 2021; 40:226920. [PMID: 33169786 PMCID: PMC7685010 DOI: 10.1042/bsr20201704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022] Open
Abstract
Skin cutaneous melanoma (SKCM) is one of the most deadly malignancies. Although immunotherapies showed the potential to improve the prognosis for metastatic melanoma patients, only a small group of patients can benefit from it. Therefore, it is urgent to investigate the tumor microenvironment in melanoma as well as to identify efficient biomarkers in the diagnosis and treatments of SKCM patients. A comprehensive analysis was performed based on metastatic melanoma samples from the Cancer Genome Atlas (TCGA) database and ESTIMATE algorithm, including gene expression, immune and stromal scores, prognostic immune‐related genes, infiltrating immune cells analysis and immune subtype identification. Then, the differentially expressed genes (DEGs) were obtained based on the immune and stromal scores, and a list of prognostic immune‐related genes was identified. Functional analysis and the protein–protein interaction network revealed that these genes enriched in multiple immune-related biological processes. Furthermore, prognostic genes were verified in the Gene Expression Omnibus (GEO) databases and used to predict immune infiltrating cells component. Our study revealed seven immune subtypes with different risk values and identified T cells as the most abundant cells in the immune microenvironment and closely associated with prognostic outcomes. In conclusion, the present study thoroughly analyzed the tumor microenvironment and identified prognostic immune‐related biomarkers for metastatic melanoma.
Collapse
|
32
|
Elsaid R, Meunier S, Burlen-Defranoux O, Soares-da-Silva F, Perchet T, Iturri L, Freyer L, Vieira P, Pereira P, Golub R, Bandeira A, Perdiguero EG, Cumano A. A wave of bipotent T/ILC-restricted progenitors shapes the embryonic thymus microenvironment in a time-dependent manner. Blood 2021; 137:1024-1036. [PMID: 33025012 PMCID: PMC8065239 DOI: 10.1182/blood.2020006779] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022] Open
Abstract
During embryonic development, multiple waves of hematopoietic progenitors with distinct lineage potential are differentially regulated in time and space. Two different waves of thymic progenitors colonize the fetal thymus where they contribute to thymic organogenesis and homeostasis. The origin, the lineage differentiation potential of the first wave, and their relative contribution in shaping the thymus architecture, remained, however, unclear. Here, we show that the first wave of thymic progenitors comprises a unique population of bipotent T and innatel lymphoid cells (T/ILC), generating a lymphoid tissue inducer cells (LTi's), in addition to invariant Vγ5+ T cells. Transcriptional analysis revealed that innate lymphoid gene signatures and, more precisely, the LTi-associated transcripts were expressed in the first, but not in the second, wave of thymic progenitors. Depletion of early thymic progenitors in a temporally controlled manner showed that the progeny of the first wave is indispensable for the differentiation of autoimmune regulator-expressing medullary thymic epithelial cells (mTECs). We further show that these progenitors are of strict hematopoietic stem cell origin, despite the overlap between lymphopoiesis initiation and the transient expression of lymphoid-associated transcripts in yolk sac (YS) erythromyeloid-restricted precursors. Our work highlights the relevance of the developmental timing on the emergence of different lymphoid subsets, required for the establishment of a functionally diverse immune system.
Collapse
Affiliation(s)
- Ramy Elsaid
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Sylvain Meunier
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Odile Burlen-Defranoux
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Francisca Soares-da-Silva
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Instituto de Investigação e Inovação em Saúde (I3S) and
- Instituto Nacional de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Thibaut Perchet
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Lorea Iturri
- Macrophages and Endothelial Cells Group, Development and Stem Cell Biology Department, Institut Pasteur, Paris, France; and
- Cellule Pasteur, University Pierre et Marie Curie (UPMC), Paris, France
| | - Laina Freyer
- Macrophages and Endothelial Cells Group, Development and Stem Cell Biology Department, Institut Pasteur, Paris, France; and
| | - Paulo Vieira
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Pablo Pereira
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Rachel Golub
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Antonio Bandeira
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Elisa Gomez Perdiguero
- Macrophages and Endothelial Cells Group, Development and Stem Cell Biology Department, Institut Pasteur, Paris, France; and
| | - Ana Cumano
- Unit of Lymphopoiesis, Immunology Department, Institut Pasteur, Paris, France
- Unité 1223, INSERM, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
33
|
Mukherjee K, Xue L, Planutis A, Gnanapragasam MN, Chess A, Bieker JJ. EKLF/KLF1 expression defines a unique macrophage subset during mouse erythropoiesis. eLife 2021; 10:61070. [PMID: 33570494 PMCID: PMC7932694 DOI: 10.7554/elife.61070] [Citation(s) in RCA: 19] [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: 07/14/2020] [Accepted: 02/10/2021] [Indexed: 12/17/2022] Open
Abstract
Erythroblastic islands are a specialized niche that contain a central macrophage surrounded by erythroid cells at various stages of maturation. However, identifying the precise genetic and transcriptional control mechanisms in the island macrophage remains difficult due to macrophage heterogeneity. Using unbiased global sequencing and directed genetic approaches focused on early mammalian development, we find that fetal liver macrophages exhibit a unique expression signature that differentiates them from erythroid and adult macrophage cells. The importance of erythroid Krüppel-like factor (EKLF)/KLF1 in this identity is shown by expression analyses in EKLF-/- and in EKLF-marked macrophage cells. Single-cell sequence analysis simplifies heterogeneity and identifies clusters of genes important for EKLF-dependent macrophage function and novel cell surface biomarkers. Remarkably, this singular set of macrophage island cells appears transiently during embryogenesis. Together, these studies provide a detailed perspective on the importance of EKLF in the establishment of the dynamic gene expression network within erythroblastic islands in the developing embryo and provide the means for their efficient isolation.
Collapse
Affiliation(s)
- Kaustav Mukherjee
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of MedicineNew York, NYUnited States
- Black Family Stem Cell InstituteNew York, NYUnited States
| | - Li Xue
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of MedicineNew York, NYUnited States
| | - Antanas Planutis
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of MedicineNew York, NYUnited States
| | - Merlin Nithya Gnanapragasam
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of MedicineNew York, NYUnited States
| | - Andrew Chess
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of MedicineNew York, NYUnited States
| | - James J Bieker
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of MedicineNew York, NYUnited States
- Black Family Stem Cell InstituteNew York, NYUnited States
- Tisch Cancer InstituteNew York, NYUnited States
- Mindich Child Health and Development Institute, Mount Sinai School of MedicineNew York, NYUnited States
| |
Collapse
|
34
|
Environmental signals rather than layered ontogeny imprint the function of type 2 conventional dendritic cells in young and adult mice. Nat Commun 2021; 12:464. [PMID: 33469015 PMCID: PMC7815729 DOI: 10.1038/s41467-020-20659-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 12/13/2020] [Indexed: 01/29/2023] Open
Abstract
Conventional dendritic cells (cDC) are key activators of naive T cells, and can be targeted in adults to induce adaptive immunity, but in early life are considered under-developed or functionally immature. Here we show that, in early life, when the immune system develops, cDC2 exhibit a dual hematopoietic origin and, like other myeloid and lymphoid cells, develop in waves. Developmentally distinct cDC2 in early life, despite being distinguishable by fate mapping, are transcriptionally and functionally similar. cDC2 in early and adult life, however, are exposed to distinct cytokine environments that shape their transcriptional profile and alter their ability to sense pathogens, secrete cytokines and polarize T cells. We further show that cDC2 in early life, despite being distinct from cDC2 in adult life, are functionally competent and can induce T cell responses. Our results thus highlight the potential of harnessing cDC2 for boosting immunity in early life.
Collapse
|
35
|
Martin EW, Krietsch J, Reggiardo RE, Sousae R, Kim DH, Forsberg EC. Chromatin accessibility maps provide evidence of multilineage gene priming in hematopoietic stem cells. Epigenetics Chromatin 2021; 14:2. [PMID: 33407811 PMCID: PMC7789351 DOI: 10.1186/s13072-020-00377-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
Hematopoietic stem cells (HSCs) have the capacity to differentiate into vastly different types of mature blood cells. The epigenetic mechanisms regulating the multilineage ability, or multipotency, of HSCs are not well understood. To test the hypothesis that cis-regulatory elements that control fate decisions for all lineages are primed in HSCs, we used ATAC-seq to compare chromatin accessibility of HSCs with five unipotent cell types. We observed the highest similarity in accessibility profiles between megakaryocyte progenitors and HSCs, whereas B cells had the greatest number of regions with de novo gain in accessibility during differentiation. Despite these differences, we identified cis-regulatory elements from all lineages that displayed epigenetic priming in HSCs. These findings provide new insights into the regulation of stem cell multipotency, as well as a resource to identify functional drivers of lineage fate.
Collapse
Affiliation(s)
- Eric W Martin
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - Jana Krietsch
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - Roman E Reggiardo
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - Rebekah Sousae
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - Daniel H Kim
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA, 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA, 95064, USA.
| |
Collapse
|
36
|
Corbin AL, Gomez-Vazquez M, Berthold DL, Attar M, Arnold IC, Powrie FM, Sansom SN, Udalova IA. IRF5 guides monocytes toward an inflammatory CD11c + macrophage phenotype and promotes intestinal inflammation. Sci Immunol 2020; 5:5/47/eaax6085. [PMID: 32444476 DOI: 10.1126/sciimmunol.aax6085] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 04/02/2020] [Indexed: 12/11/2022]
Abstract
Mononuclear phagocytes (MNPs) are vital for maintaining intestinal homeostasis but, in response to acute microbial stimulation, can also trigger immunopathology, accelerating recruitment of Ly6Chi monocytes to the gut. The regulators that control monocyte tissue adaptation in the gut remain poorly understood. Interferon regulatory factor 5 (IRF5) is a transcription factor previously shown to play a key role in maintaining the inflammatory phenotype of macrophages. Here, we investigate the impact of IRF5 on the MNP system and physiology of the gut at homeostasis and during inflammation. We demonstrate that IRF5 deficiency has a limited impact on colon physiology at steady state but ameliorates immunopathology during Helicobacter hepaticus-induced colitis. Inhibition of IRF5 activity in MNPs phenocopies global IRF5 deficiency. Using a combination of bone marrow chimera and single-cell RNA-sequencing approaches, we examined the intrinsic role of IRF5 in controlling colonic MNP development. We demonstrate that IRF5 promotes differentiation of Ly6Chi monocytes into CD11c+ macrophages and controls the production of antimicrobial and inflammatory mediators by these cells. Thus, we identify IRF5 as a key transcriptional regulator of the colonic MNP system during intestinal inflammation.
Collapse
Affiliation(s)
| | | | | | - Moustafa Attar
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Isabelle C Arnold
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.,Institut für Molekulare Krebsforschung, University of Zurich, Zurich, Switzerland
| | - Fiona M Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Stephen N Sansom
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| |
Collapse
|
37
|
Evren E, Ringqvist E, Tripathi KP, Sleiers N, Rives IC, Alisjahbana A, Gao Y, Sarhan D, Halle T, Sorini C, Lepzien R, Marquardt N, Michaëlsson J, Smed-Sörensen A, Botling J, Karlsson MCI, Villablanca EJ, Willinger T. Distinct developmental pathways from blood monocytes generate human lung macrophage diversity. Immunity 2020; 54:259-275.e7. [PMID: 33382972 DOI: 10.1016/j.immuni.2020.12.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/15/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.
Collapse
Affiliation(s)
- Elza Evren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Emma Ringqvist
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Kumar Parijat Tripathi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Natalie Sleiers
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Inés Có Rives
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Arlisa Alisjahbana
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Yu Gao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Tor Halle
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Chiara Sorini
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Rico Lepzien
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Nicole Marquardt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Tim Willinger
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden.
| |
Collapse
|
38
|
Cool T, Worthington A, Poscablo D, Hussaini A, Forsberg EC. Interleukin 7 receptor is required for myeloid cell homeostasis and reconstitution by hematopoietic stem cells. Exp Hematol 2020; 90:39-45.e3. [PMID: 32916215 DOI: 10.1016/j.exphem.2020.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 12/24/2022]
Abstract
Respiratory diseases are a leading cause of death worldwide, with vulnerability to disease varying greatly between individuals. The reasons underlying disease susceptibility are unknown, but there is often a variable immune response in lungs often. Recently, we identified a surprising novel role for the interleukin 7 receptor (IL7R), a primarily lymphoid-associated regulator, in fetal-specified, lung-resident macrophage development. Here, we report that traditional, hematopoietic stem cell-derived myeloid cells in the adult lung, peripheral blood, and bone marrow also depend on IL7R expression. Using single- and double-germline knockout models, we found that eosinophil numbers were reduced on deletion of IL7Rα. We then employed two Cre recombinase models in lineage tracing experiments to test whether these cells developed through an IL7Rα+ pathway. Despite the impact of IL7Rα deletion, IL7R-Cre labeled only a minimal fraction of eosinophils. We therefore examined the intrinsic versus extrinsic requirement for IL7R in the production of eosinophils using reciprocal hematopoietic stem cell transplantation assays. These assays revealed that extrinsic, but not eosinophil-intrinsic, IL7R is required for eosinophil reconstitution by HSCs in the adult lung. To determine which external factors may be influencing eosinophil development and survival, we performed a cytokine array analysis between wild-type and IL7Rα-deficient mice and found several differentially regulated proteins. These findings expand on our previous report that IL7R is required not only for proper lymphoid cell development and homeostasis, but also for myeloid cell homeostasis in tissues.
Collapse
Affiliation(s)
- Taylor Cool
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA
| | - Atesh Worthington
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA
| | - Donna Poscablo
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; Program in Biomedical Sciences and Engineering, Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA
| | - Adeel Hussaini
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; Biomolecular Engineering, University of California-Santa Cruz, Santa Cruz, CA.
| |
Collapse
|
39
|
Li Z, Cui Y, Feng J, Guo Y. Identifying the pattern of immune related cells and genes in the peripheral blood of ischemic stroke. J Transl Med 2020; 18:296. [PMID: 32746852 PMCID: PMC7398186 DOI: 10.1186/s12967-020-02463-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 07/28/2020] [Indexed: 12/31/2022] Open
Abstract
Background Ischemic stroke (IS) is the second leading cause of death worldwide which is a serious hazard to human health. Evidence suggests that the immune system plays a key role in the pathophysiology of IS. However, the precisely immune related mechanisms were still not been systematically understood. Methods In this study, we aim to identify the immune related modules and genes that might play vital role in the occurrence and development of IS by using the weighted gene co-expression network analysis (WGCNA). Meanwhile, we applied a kind of deconvolution algorithm to reveal the proportions of 22 subsets of immune cells in the blood samples. Results There were total 128 IS patients and 67 healthy control samples in the three Gene Expression Omnibus (GEO) datasets. Under the screening criteria, 1082 DEGs (894 up-regulated and 188 down-regulated) were chosen for further analysis. A total of 11 clinically significant modules were identified, from which immune-related hub modules and hub genes were further explored. Finally, 16 genes were selected as real hub genes for further validation analysis. Furthermore, these CIBERSORT results suggest that detailed analysis of the immune subtype distribution pattern has the potential to enhance clinical prediction and to identify candidates for immunotherapy. More specifically, we identified that neutrophil emerge as a promising target for IS therapies. Conclusions In the present study, we investigated the immune related gene expression modules, in which the SLAMF1, IL7R and NCF4 may be novel therapeutic targets to promote functional and histological recovery after ischemic stroke. Furthermore, these hub genes and neutrophils may become important biological targets in the drug screening and drug designing.
Collapse
Affiliation(s)
- Zijian Li
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, People's Republic of China
| | - Yueran Cui
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, People's Republic of China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, People's Republic of China
| | - Yanxia Guo
- Department of Neurology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, People's Republic of China.
| |
Collapse
|
40
|
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
|
41
|
Coakley G, Volpe B, Bouchery T, Shah K, Butler A, Geldhof P, Hatherill M, Horsnell WGC, Esser-von Bieren J, Harris NL. Immune serum-activated human macrophages coordinate with eosinophils to immobilize Ascaris suum larvae. Parasite Immunol 2020; 42:e12728. [PMID: 32394439 DOI: 10.1111/pim.12728] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/19/2022]
Abstract
Helminth infection represents a major health problem causing approximately 5 million disability-adjusted life years worldwide. Concerns that repeated anti-helminthic treatment may lead to drug resistance render it important that vaccines are developed but will require increased understanding of the immune-mediated cellular and antibody responses to helminth infection. IL-4 or antibody-activated murine macrophages are known to immobilize parasitic nematode larvae, but few studies have addressed whether this is translatable to human macrophages. In the current study, we investigated the capacity of human macrophages to recognize and attack larval stages of Ascaris suum, a natural porcine parasite that is genetically similar to the human helminth Ascaris lumbricoides. Human macrophages were able to adhere to and trap A suum larvae in the presence of either human or pig serum containing Ascaris-specific antibodies and other factors. Gene expression analysis of serum-activated macrophages revealed that CCL24, a potent eosinophil attractant, was the most upregulated gene following culture with A suum larvae in vitro, and human eosinophils displayed even greater ability to adhere to, and trap, A suum larvae. These data suggest that immune serum-activated macrophages can recruit eosinophils to the site of infection, where they act in concert to immobilize tissue-migrating Ascaris larvae.
Collapse
Affiliation(s)
- Gillian Coakley
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Beatrice Volpe
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Tiffany Bouchery
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Kathleen Shah
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Alana Butler
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Peter Geldhof
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Mark Hatherill
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - William G C Horsnell
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Julia Esser-von Bieren
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland.,Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Nicola Laraine Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| |
Collapse
|
42
|
Kubin N, Richter M, Sen-Hild B, Akintürk H, Schönburg M, Kubin T, Cetinkaya A. Macrophages represent the major pool of IL-7Rα expressing cells in patients with myocarditis. Cytokine 2020; 130:155053. [PMID: 32203694 DOI: 10.1016/j.cyto.2020.155053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/02/2020] [Accepted: 02/24/2020] [Indexed: 11/15/2022]
Abstract
Myocarditis is characterized by infiltration and activation of cytokine as well as chemokine receptors frequently producing heart failure. Causes are often infections triggering inflammatory and immune responses but these initial lines of defense might be finally disastrous. To identify mediators we screened various receptors by confocal microscopy and identified cardiac interleukin-7 (IL-7) receptor-α (IL-7Rα) expressing cells in patients with myocarditis. IL-7Rα+ cells were analyzed by markers for leukocytes (CD45), B cells (CD19), T cells (CD3, CD4, CD8) and macrophages (CD68, CD163, CD206). Immune cells were hardly detected in controls. In patients with myocarditis main inflammatory populations consisted of macrophages and T cells. B cells were hardly present. 90% of CD68+ macrophages but less than 20% of CD3+ T cells were IL-7Rα+. This was surprising since T and B lymphocytes are generally regarded as the major IL-7Rα+ cells. Since IL-7 acts as a chemokine, the expression of its receptor might orchestrate cardiac macrophage infiltration. In contrast, consumption of IL-7 by IL-7Rα+ cardiac macrophages might potentially prevent a certain overshooting immune reaction and sepsis by reducing proliferation and survival of lymphocytes. Our data suggest a participation of IL-7Rα+ macrophages in the development of myocarditis and heart failure.
Collapse
Affiliation(s)
- Natalia Kubin
- Department of Cardiac Surgery, Kerckhoff-Clinic, Benekestrasse 2-8, Bad Nauheim 61231, Germany
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Clinic, Benekestrasse 2-8, Bad Nauheim 61231, Germany.
| | - Bedriye Sen-Hild
- Pediatric Heart Center, Justus Liebig University, Feulgenstrasse 10-12, Giessen 35392, Germany
| | - Hakan Akintürk
- Pediatric Heart Center, Justus Liebig University, Feulgenstrasse 10-12, Giessen 35392, Germany
| | - Markus Schönburg
- Department of Cardiac Surgery, Kerckhoff-Clinic, Benekestrasse 2-8, Bad Nauheim 61231, Germany
| | - Thomas Kubin
- Department of Cardiac Surgery, Kerckhoff-Clinic, Benekestrasse 2-8, Bad Nauheim 61231, Germany
| | - Ayse Cetinkaya
- Department of Cardiac Surgery, Kerckhoff-Clinic, Benekestrasse 2-8, Bad Nauheim 61231, Germany
| |
Collapse
|
43
|
Luo S, Zhu R, Yu T, Fan H, Hu Y, Mohanta SK, Hu D. Chronic Inflammation: A Common Promoter in Tertiary Lymphoid Organ Neogenesis. Front Immunol 2019; 10:2938. [PMID: 31921189 PMCID: PMC6930186 DOI: 10.3389/fimmu.2019.02938] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 11/29/2019] [Indexed: 12/15/2022] Open
Abstract
Tertiary lymphoid organs (TLOs) frequently develop locally in adults in response to non-resolving inflammation. Chronic inflammation leads to the differentiation of stromal fibroblast cells toward lymphoid tissue organizer-like cells, which interact with lymphotoxin α1β2+ immune cells. The interaction initiates lymphoid neogenesis by recruiting immune cells to the site of inflammation and ultimately leads to the formation of TLOs. Mature TLOs harbor a segregated T-cell zone, B-cell follicles with an activated germinal center, follicular dendritic cells, and high endothelial venules, which architecturally resemble those in secondary lymphoid organs. Since CXCL13 and LTα1β2 play key roles in TLO neogenesis, they might constitute potential biomarkers of TLO activity. The well-developed TLOs actively regulate local immune responses and influence disease progression, and they are thereby regarded as the powerhouses of local immunity. In this review, we recapitulated the determinants for TLOs development, with great emphasis on the fundamental role of chronic inflammation and tissue-resident stromal cells for TLO neogenesis, hence offering guidance for therapeutic interventions in TLO-associated diseases.
Collapse
Affiliation(s)
- Shanshan Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Yu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sarajo Kumar Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Desheng Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Flip the coin: IL-7 and IL-7R in health and disease. Nat Immunol 2019; 20:1584-1593. [PMID: 31745336 DOI: 10.1038/s41590-019-0479-x] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022]
Abstract
The cytokine IL-7 and its receptor, IL-7R, are critical for T cell and, in the mouse, B cell development, as well as differentiation and survival of naive T cells, and generation and maintenance of memory T cells. They are also required for innate lymphoid cell (ILC) development and maintenance, and consequently for generation of lymphoid structures and barrier defense. Here we discuss the central role of IL-7 and IL-7R in the lymphoid system and highlight the impact of their deregulation, placing a particular emphasis on their 'dark side' as promoters of cancer development. We also explore therapeutic implications and opportunities associated with either positive or negative modulation of the IL-7-IL-7R signaling axis.
Collapse
|
45
|
Smith-Berdan S, Bercasio A, Rajendiran S, Forsberg EC. Viagra Enables Efficient, Single-Day Hematopoietic Stem Cell Mobilization. Stem Cell Reports 2019; 13:787-792. [PMID: 31607567 PMCID: PMC6895718 DOI: 10.1016/j.stemcr.2019.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 09/10/2019] [Accepted: 09/10/2019] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation is a curative treatment for a variety of blood and immune disorders. Currently available methods to obtain donor HSCs are suboptimal, and the limited supply of donor HSCs hampers the success and availability of HSC transplantation therapies. We recently showed that manipulation of vascular integrity can be employed to induce HSC mobilization from the bone marrow to the blood stream, facilitating non-invasive collection of HSCs. Here, we tested whether FDA-approved vasodilators are capable of mobilizing HSCs. We found that a rapid, 2-h regimen of a single oral dose of Viagra (sildenafil citrate) combined with a single injection of the CXCR4 antagonist AMD3100 leads to efficient HSC mobilization at levels rivaling the standard-of-care 5-day regimen of granulocyte-colony stimulating factor (G-CSF/Filgrastim/Neupogen). Our findings solidify vascular integrity as an essential regulator of HSC trafficking and provide an attractive, single-day regimen for HSC mobilization using already FDA-approved drugs. Viagra enhances AMD3100-mediated HSC mobilization Extremely rapid and efficient HSC mobilization with two FDA-approved drugs Vascular integrity regulates HSC trafficking
Collapse
Affiliation(s)
- Stephanie Smith-Berdan
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Alyssa Bercasio
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA 95064, USA
| | - Smrithi Rajendiran
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA 95064, USA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, 1156 High Street, Santa Cruz, CA 95064, USA.
| |
Collapse
|
46
|
Wang L, Wei CY, Xu YY, Deng XY, Wang Q, Ying JH, Zhang SM, Yuan X, Xuan TF, Pan YY, Gu JY. Prognostic genes of melanoma identified by weighted gene co-expression network analysis and drug repositioning using a network-based method. Oncol Lett 2019; 18:6066-6078. [PMID: 31788081 PMCID: PMC6864934 DOI: 10.3892/ol.2019.10961] [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: 02/12/2019] [Accepted: 08/21/2019] [Indexed: 12/19/2022] Open
Abstract
Melanoma is one of the most malignant types of skin cancer. However, the efficacy and utility of available drug therapies for melanoma are limited. The objective of the present study was to identify potential genes associated with melanoma progression and to explore approved therapeutic drugs that target these genes. Weighted gene co-expression network analysis was used to construct a gene co-expression network, explore the associations between genes and clinical characteristics and identify potential biomarkers. Gene expression profiles of the GSE65904 dataset were obtained from the Gene Expression Omnibus database. RNA-sequencing data and clinical information associated with melanoma obtained from The Cancer Genome Atlas were used for biomarker validation. A total of 15 modules were identified through average linkage hierarchical clustering. In the two significant modules, three network hub genes associated with melanoma prognosis were identified: C-X-C motif chemokine receptor 4 (CXCR4), interleukin 7 receptor (IL7R) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit γ (PIK3CG). The receiver operating characteristic curve indicated that the mRNA levels of these genes exhibited excellent prognostic efficiency for primary and metastatic tumor tissues. In addition, the proximity between candidate genes associated with melanoma progression and drug targets obtained from DrugBank was calculated in the protein interaction network, and the top 15 drugs that may be suitable for treating melanoma were identified. In summary, co-expression network analysis led to the selection of CXCR4, IL7R and PIK3CG for further basic and clinical research on melanoma. Utilizing a network-based method, 15 drugs that exhibited potential for the treatment of melanoma were identified.
Collapse
Affiliation(s)
- Lu Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Chuan-Yuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yuan-Yuan Xu
- Department of Surgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xin-Yi Deng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Qiang Wang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jiang-Hui Ying
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Si-Min Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xin Yuan
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Tian-Fan Xuan
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Yu-Yan Pan
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jian-Ying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|