1
|
Khalaf K, Chamieh M, Welc N, Singh C, Kaouk JL, Kaouk A, Mackiewicz A, Kaczmarek M, Perek B. Cellular aspects of immunity involved in the development of atherosclerosis. Front Immunol 2025; 16:1461535. [PMID: 39944697 PMCID: PMC11813763 DOI: 10.3389/fimmu.2025.1461535] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 01/09/2025] [Indexed: 05/09/2025] Open
Abstract
Atherosclerosis, previously regarded as a lipid storage disease, has now been classified as a chronic inflammatory disease. The hardening of arterial vessels characterizes atherosclerosis due to the accumulation of lipids in the arterial walls, eliciting an inflammatory response. The development of atherosclerosis occurs in various stages and is facilitated by many clinical factors, such as hypertension, hyperlipidemia, and inflammatory status. A large arsenal of cells has been implicated in its development. This review will summarize the phases of atherosclerotic formation and all the cells involved in either promoting or inhibiting its development.
Collapse
Affiliation(s)
- Khalil Khalaf
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| | - Marc Chamieh
- Department of Spine Disorders and Pediatric Orthopedics, Poznan University of Medical Sciences, Poznań, Poland
| | - Natalia Welc
- Department of Dermatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Chandpreet Singh
- Department of Internal Medicine, University of California, Los Angeles (UCLA) - Kern Medical Center, Bakersfield, CA, United States
| | - Joanne Lynn Kaouk
- Department of Science, Louisiana State University, Lousiana, LA, United States
| | - Aiden Kaouk
- Department of Natural Sciences, The University of Texas at Austin, Texas, TX, United States
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
| | - Bartlomiej Perek
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
2
|
Ma Z, Liu L, Tian J, Tu C, Zhang D, Zhang M, Zhang H, An Z, Sun M, Zhang H, Song X. Causal Relationships between Lymphocyte Subsets and Risk of Coronary Artery Disease: A Two-Sample Mendelian Randomization Study. Rev Cardiovasc Med 2024; 25:326. [PMID: 39355583 PMCID: PMC11440411 DOI: 10.31083/j.rcm2509326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 10/03/2024] Open
Abstract
Background Currently, the causal relationship between lymphocyte subsets and coronary artery disease (CAD) remains unclear. Therefore, we utilized Mendelian randomization (MR) to assess the association between lymphocyte subsets and CAD. Methods We performed a two-sample MR analysis using publicly available genome-wide association studies (GWAS) datasets. The primary method of analysis to comprehensively evaluate causal effects was the inverse variance-weighted (IVW) method. The four additional MR approaches were MR-Egger, weighted median, simple mode, and weighted mode. Sensitivity analysis incorporated Cochran's Q and MR-Egger intercept tests to identify residual heterogeneity and potential horizontal pleiotropy, respectively. The MR-PRESSO distortion test was applied to identify potential pleiotropic outliers. Leave-one-out analysis confirmed that no single single-nucleotide polymorphism (SNP) significantly affected the MR estimate. We conducted reverse MR analysis to investigate the impact of variables correlated with outcomes in forward MR analysis. Results The IVW method revealed a significant positive association between B cell count and CAD (odds ratio (OR) = 1.08 (95% CI: 1.04, 1.11), p = 2.67 × 10-5). A similar association was observed between B cell count and myocardial infarction (MI) (OR = 1.07 (95% CI: 1.03, 1.11), p = 5.69 × 10-4). Sensitivity analyses detected no outliers, heterogeneity, or pleiotropy. The reverse MR analysis was conducted to investigate the impact of CAD and MI on B cell count, and the IVW results showed no statistical significance. Conclusions Our study suggests that a higher absolute B cell count is linked to an increased risk of CAD and MI.
Collapse
Affiliation(s)
- Zhao Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Chenchen Tu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Dongfeng Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Mingduo Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Meichen Sun
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Hongjia Zhang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| |
Collapse
|
3
|
Meher AK, McNamara CA. B-1 lymphocytes in adipose tissue as innate modulators of inflammation linked to cardiometabolic disease. Immunol Rev 2024; 324:95-103. [PMID: 38747455 PMCID: PMC11262958 DOI: 10.1111/imr.13342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Fat is stored in distinct depots with unique features in both mice and humans and B cells reside in all adipose depots. We have shown that B cells modulate cardiometabolic disease through activities in two of these key adipose depots: visceral adipose tissue (VAT) and perivascular adipose tissue (PVAT). VAT refers to the adipose tissue surrounding organs, within the abdomen and thorax, and is comprised predominantly of white adipocytes. This depot has been implicated in mediating obesity-related dysmetabolism. PVAT refers to adipose tissue surrounding major arteries. It had long been thought to exist to provide protection and insulation for the vessel, yet recent work demonstrates an important role for PVAT in harboring immune cells, promoting their function and regulating the biology of the underlying vessel. The role of B-2 cells and adaptive immunity in adipose tissue biology has been nicely reviewed elsewhere. Given that, the predominance of B-1 cells in adipose tissue at homeostasis, and the emerging role of B-1 cells in a variety of disease states, we will focus this review on how B-1 cells function in VAT and PVAT depots to promote homeostasis and limit inflammation linked to cardiometabolic disease and factors that regulate this function.
Collapse
Affiliation(s)
- Akshaya K. Meher
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Coleen A. McNamara
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
4
|
Cheng T, You Y, Jia B, Wang H, Lv M, Zhu X, Hu Y. Knowledge mapping of B cell and atherosclerosis over the past 20 years: A bibliometric analysis. Hum Vaccin Immunother 2023; 19:2277567. [PMID: 37953301 PMCID: PMC10760366 DOI: 10.1080/21645515.2023.2277567] [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: 08/18/2023] [Accepted: 10/27/2023] [Indexed: 11/14/2023] Open
Abstract
Atherosclerosis (AS) is the main underlying cause of cardiovascular disease, and B cells are considered a key immune cell type to regulate AS. So far, there is no bibliometric study on B cell and AS. This study aims to comprehensively analyze the scientific output about B cell and AS, summarize the literature characteristics, explore research hotspots, and point out emerging trends. We searched the literature from 2003 to 2022 from the Web of Science Core Collection (WoSCC) database. CiteSpace, VOSviewer, and the R package "Bibliometrix" were used for literature analysis and visualization. A total of 1,062 articles and reviews were identified. The number of annual publications generally showed an upward trend. The United States and China were the most productive countries. Medical University of Vienna was the most productive research institution, and Binder Christoph J. was the most productive author, who was also from Medical University of Vienna. "Arteriosclerosis Thrombosis and Vascular Biology" was the most published journal and the most frequently cited journal. The most cited reference was written by Caligiuri G (2002) in "Journal of Clinical Investigation." The most frequent keywords were "inflammation," "macrophages," "cardiovascular disease," "T cells," "apoptosis," "immunity," "cytokines," "lymphocytes," etc. The trend topics were mainly focused on "immune infiltration," "immunoglobulins," and "biomarkers." The complex role of B cell subtypes and a variety of B cell mediators is the main research direction at present. In-depth analysis of B cell-specific targets can provide new ideas and methods for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Tao Cheng
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Clinical Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yaping You
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Bochao Jia
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
- Clinical Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Huan Wang
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Meng Lv
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Xueping Zhu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| | - Yuanhui Hu
- Department of Cardiological Medicine, China Academy of Chinese Medical Sciences Guang’anmen Hospital, Beijing, China
| |
Collapse
|
5
|
Sun S, Wijanarko K, Liani O, Strumila K, Ng ES, Elefanty AG, Stanley EG. Lymphoid cell development from fetal hematopoietic progenitors and human pluripotent stem cells. Immunol Rev 2023; 315:154-170. [PMID: 36939073 PMCID: PMC10952469 DOI: 10.1111/imr.13197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
Lymphoid cells encompass the adaptive immune system, including T and B cells and Natural killer T cells (NKT), and innate immune cells (ILCs), including Natural Killer (NK) cells. During adult life, these lineages are thought to derive from the differentiation of long-term hematopoietic stem cells (HSCs) residing in the bone marrow. However, during embryogenesis and fetal development, the ontogeny of lymphoid cells is both complex and multifaceted, with a large body of evidence suggesting that lymphoid lineages arise from progenitor cell populations antedating the emergence of HSCs. Recently, the application of single cell RNA-sequencing technologies and pluripotent stem cell-based developmental models has provided new insights into lymphoid ontogeny during embryogenesis. Indeed, PSC differentiation platforms have enabled de novo generation of lymphoid immune cells independently of HSCs, supporting conclusions drawn from the study of hematopoiesis in vivo. Here, we examine lymphoid development from non-HSC progenitor cells and technological advances in the differentiation of human lymphoid cells from pluripotent stem cells for clinical translation.
Collapse
Affiliation(s)
- Shicheng Sun
- Murdoch Children's Research InstituteThe Royal Children's HospitalParkvilleVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Kevin Wijanarko
- Murdoch Children's Research InstituteThe Royal Children's HospitalParkvilleVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Oniko Liani
- Murdoch Children's Research InstituteThe Royal Children's HospitalParkvilleVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Kathleen Strumila
- Murdoch Children's Research InstituteThe Royal Children's HospitalParkvilleVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Elizabeth S. Ng
- Murdoch Children's Research InstituteThe Royal Children's HospitalParkvilleVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Andrew G. Elefanty
- Murdoch Children's Research InstituteThe Royal Children's HospitalParkvilleVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| | - Edouard G. Stanley
- Murdoch Children's Research InstituteThe Royal Children's HospitalParkvilleVictoriaAustralia
- Department of Paediatrics, Faculty of Medicine, Dentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Murdoch Children's Research InstituteParkvilleVictoriaAustralia
| |
Collapse
|
6
|
Mendoza-Castrejon J, Magee JA. Layered immunity and layered leukemogenicity: Developmentally restricted mechanisms of pediatric leukemia initiation. Immunol Rev 2023; 315:197-215. [PMID: 36588481 PMCID: PMC10301262 DOI: 10.1111/imr.13180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Hematopoietic stem cells (HSCs) and multipotent progenitor cells (MPPs) arise in successive waves during ontogeny, and their properties change significantly throughout life. Ontological changes in HSCs/MPPs underlie corresponding changes in mechanisms of pediatric leukemia initiation. As HSCs and MPPs progress from fetal to neonatal, juvenile and adult stages of life, they undergo transcriptional and epigenetic reprogramming that modifies immune output to meet age-specific pathogenic challenges. Some immune cells arise exclusively from fetal HSCs/MPPs. We propose that this layered immunity instructs cell fates that underlie a parallel layered leukemogenicity. Indeed, some pediatric leukemias, such as juvenile myelomonocytic leukemia, myeloid leukemia of Down syndrome, and infant pre-B-cell acute lymphoblastic leukemia, are age-restricted. They only present during infancy or early childhood. These leukemias likely arise from fetal progenitors that lose competence for transformation as they age. Other childhood leukemias, such as non-infant pre-B-cell acute lymphoblastic leukemia and acute myeloid leukemia, have mutation profiles that are common in childhood but rare in morphologically similar adult leukemias. These differences could reflect temporal changes in mechanisms of mutagenesis or changes in how progenitors respond to a given mutation at different ages. Interactions between leukemogenic mutations and normal developmental switches offer potential targets for therapy.
Collapse
Affiliation(s)
- Jonny Mendoza-Castrejon
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
| | - Jeffrey A. Magee
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
| |
Collapse
|
7
|
Kobayashi M, Yoshimoto M. Multiple waves of fetal-derived immune cells constitute adult immune system. Immunol Rev 2023; 315:11-30. [PMID: 36929134 PMCID: PMC10754384 DOI: 10.1111/imr.13192] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
It has been over three decades since Drs. Herzenberg and Herzenberg proposed the layered immune system hypothesis, suggesting that different types of stem cells with distinct hematopoietic potential produce specific immune cells. This layering of immune system development is now supported by recent studies showing the presence of fetal-derived immune cells that function in adults. It has been shown that various immune cells arise at different embryonic ages via multiple waves of hematopoiesis from special endothelial cells (ECs), referred to as hemogenic ECs. However, it remains unknown whether these fetal-derived immune cells are produced by hematopoietic stem cells (HSCs) during the fetal to neonatal period. To address this question, many advanced tools have been used, including lineage-tracing mouse models, cellular barcoding techniques, clonal assays, and transplantation assays at the single-cell level. In this review, we will review the history of the search for the origins of HSCs, B-1a progenitors, and mast cells in the mouse embryo. HSCs can produce both B-1a and mast cells within a very limited time window, and this ability declines after embryonic day (E) 14.5. Furthermore, the latest data have revealed that HSC-independent adaptive immune cells exist in adult mice, which implies more complicated developmental pathways of immune cells. We propose revised road maps of immune cell development.
Collapse
Affiliation(s)
- Michihiro Kobayashi
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Momoko Yoshimoto
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
8
|
Hiéronimus L, Huaux F. B-1 cells in immunotoxicology: Mechanisms underlying their response to chemicals and particles. FRONTIERS IN TOXICOLOGY 2023; 5:960861. [PMID: 37143777 PMCID: PMC10151831 DOI: 10.3389/ftox.2023.960861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
Since their discovery nearly 40 years ago, B-1 cells have continued to challenge the boundaries between innate and adaptive immunity, as well as myeloid and lymphoid functions. This B-cell subset ensures early immunity in neonates before the development of conventional B (B-2) cells and respond to immune injuries throughout life. B-1 cells are multifaceted and serve as natural- and induced-antibody-producing cells, phagocytic cells, antigen-presenting cells, and anti-/pro-inflammatory cytokine-releasing cells. This review retraces the origin of B-1 cells and their different roles in homeostatic and infectious conditions before focusing on pollutants comprising contact-sensitivity-inducing chemicals, endocrine disruptors, aryl hydrocarbon receptor (AHR) ligands, and reactive particles.
Collapse
|
9
|
Olson WJ, Jakic B, Labi V, Woelk J, Derudder E, Baier G, Hermann-Kleiter N. A role for the nuclear receptor NR2F6 in peritoneal B cell homeostasis. Front Immunol 2022; 13:845235. [PMID: 36052079 PMCID: PMC9425112 DOI: 10.3389/fimmu.2022.845235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 07/18/2022] [Indexed: 11/19/2022] Open
Abstract
B cells are key mediators of humoral immunity. Mature B cells fall into various sub-classes that can be separated by their ontogeny, expression of cell surface markers, anatomical location, and function. B1 subsets play important roles in natural immunity and constitute the majority of B cells in newborns. In the adult, B1 cells predominate in the pleural and peritoneal cavities, while the mature B2 follicular subset makes up the major fraction of B cells in lymphoid tissue, although important subsets of antibody-secreting B1 cells are also present at these sites. B1 cells are the main producers of natural IgM but can also contribute to elimination of some pathogens, while B2 cells primarily mediate response to foreign antigens. The differential molecular underpinning of the B1 and B2 subsets remains incompletely understood. Here we demonstrate that germline-deficiency of the orphan nuclear receptor NR2F6 causes a partial loss of B1b and B2 B cells in the peritoneum while leaving peritoneal B1a cells unaltered. A competitive bone marrow chimera in Nr2f6+/+ host mice produced similar numbers of Nr2f6+/+ and Nr2f6-/- peritoneal B1b and B2 cells. The proliferation of Nr2f6-/- peritoneal B cells was not altered, while the migration marker CXCR5 was reduced on all subsets but Beta7-integrin was reduced only on peritoneal B1b and B2 cells. Similarly, B1b and B2 but not B1a cells, exhibited significantly reduced survival.
Collapse
Affiliation(s)
- William J. Olson
- Translational Cell Genetics, Department of Pharmacology and Genetics, Medical University of Innsbruck, Innsbruck, Austria
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Bojana Jakic
- Translational Cell Genetics, Department of Pharmacology and Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Verena Labi
- Institute of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Woelk
- Translational Cell Genetics, Department of Pharmacology and Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Emmanuel Derudder
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Gottfried Baier
- Translational Cell Genetics, Department of Pharmacology and Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Translational Cell Genetics, Department of Pharmacology and Genetics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
10
|
Embryonic Origins of the Hematopoietic System: Hierarchies and Heterogeneity. Hemasphere 2022; 6:e737. [PMID: 35647488 PMCID: PMC9132533 DOI: 10.1097/hs9.0000000000000737] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 04/28/2022] [Indexed: 11/26/2022] Open
Abstract
The hierarchical framework of the adult blood system as we know it from current medical and hematology textbooks, displays a linear branching network of dividing and differentiated cells essential for the growth and maintenance of the healthy organism. This view of the hierarchy has evolved over the last 75 years. An amazing increase in cellular complexity has been realized; however, innovative single-cell technologies continue to uncover essential cell types and functions in animal models and the human blood system. The most potent cell of the hematopoietic hierarchy is the hematopoietic stem cell. Stem cells for adult tissues are the long-lived self-renewing cellular component, which ensure that differentiated tissue-specific cells are maintained and replaced through the entire adult lifespan. Although much blood research is focused on hematopoietic tissue homeostasis, replacement and regeneration during adult life, embryological studies have widened and enriched our understanding of additional developmental hierarchies and interacting cells of this life-sustaining tissue. Here, we review the current state of knowledge of the hierarchical organization and the vast heterogeneity of the hematopoietic system from embryonic to adult stages.
Collapse
|
11
|
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
|
12
|
Watt SM, Hua P, Roberts I. Increasing Complexity of Molecular Landscapes in Human Hematopoietic Stem and Progenitor Cells during Development and Aging. Int J Mol Sci 2022; 23:3675. [PMID: 35409034 PMCID: PMC8999121 DOI: 10.3390/ijms23073675] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 02/05/2023] Open
Abstract
The past five decades have seen significant progress in our understanding of human hematopoiesis. This has in part been due to the unprecedented development of advanced technologies, which have allowed the identification and characterization of rare subsets of human hematopoietic stem and progenitor cells and their lineage trajectories from embryonic through to adult life. Additionally, surrogate in vitro and in vivo models, although not fully recapitulating human hematopoiesis, have spurred on these scientific advances. These approaches have heightened our knowledge of hematological disorders and diseases and have led to their improved diagnosis and therapies. Here, we review human hematopoiesis at each end of the age spectrum, during embryonic and fetal development and on aging, providing exemplars of recent progress in deciphering the increasingly complex cellular and molecular hematopoietic landscapes in health and disease. This review concludes by highlighting links between chronic inflammation and metabolic and epigenetic changes associated with aging and in the development of clonal hematopoiesis.
Collapse
Affiliation(s)
- Suzanne M. Watt
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9BQ, UK
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5005, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5001, Australia
| | - Peng Hua
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China;
| | - Irene Roberts
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, and NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK;
- Department of Paediatrics and NIHR Oxford Biomedical Research Centre Haematology Theme, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
13
|
Barone C, Orsenigo R, Meneveri R, Brunelli S, Azzoni E. One Size Does Not Fit All: Heterogeneity in Developmental Hematopoiesis. Cells 2022; 11:1061. [PMID: 35326511 PMCID: PMC8947200 DOI: 10.3390/cells11061061] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/06/2023] Open
Abstract
Our knowledge of the complexity of the developing hematopoietic system has dramatically expanded over the course of the last few decades. We now know that, while hematopoietic stem cells (HSCs) firmly reside at the top of the adult hematopoietic hierarchy, multiple HSC-independent progenitor populations play variegated and fundamental roles during fetal life, which reflect on adult physiology and can lead to disease if subject to perturbations. The importance of obtaining a high-resolution picture of the mechanisms by which the developing embryo establishes a functional hematopoietic system is demonstrated by many recent indications showing that ontogeny is a primary determinant of function of multiple critical cell types. This review will specifically focus on exploring the diversity of hematopoietic stem and progenitor cells unique to embryonic and fetal life. We will initially examine the evidence demonstrating heterogeneity within the hemogenic endothelium, precursor to all definitive hematopoietic cells. Next, we will summarize the dynamics and characteristics of the so-called "hematopoietic waves" taking place during vertebrate development. For each of these waves, we will define the cellular identities of their components, the extent and relevance of their respective contributions as well as potential drivers of heterogeneity.
Collapse
Affiliation(s)
| | | | | | | | - Emanuele Azzoni
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy; (C.B.); (R.O.); (R.M.); (S.B.)
| |
Collapse
|
14
|
Eddins DJ, Kosters A, Waters J, Sosa J, Phillips M, Yadava K, Herzenberg LA, Kuipers HF, Ghosn EEB. Hematopoietic Stem Cell Requirement for Macrophage Regeneration Is Tissue Specific. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:3028-3037. [PMID: 34810224 DOI: 10.4049/jimmunol.2100344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022]
Abstract
Tissue-resident macrophages (TRMΦ) are important immune sentinels responsible for maintaining tissue and immune homeostasis within their specific niche. Recently, the origins of TRMΦ have undergone intense scrutiny, in which now most TRMΦ are thought to originate early during embryonic development independent of hematopoietic stem cells (HSCs). We previously characterized two distinct subsets of mouse peritoneal cavity macrophages (MΦ) (large and small peritoneal MΦ) whose origins and relationship to both fetal and adult long-term (LT) HSCs have not been fully investigated. In this study, we employ highly purified LT-HSC transplantation and in vivo lineage tracing to show a dual ontogeny for large and small peritoneal MΦ, in which the initial wave of peritoneal MΦ is seeded from yolk sac-derived precursors, which later require LT-HSCs for regeneration. In contrast, transplanted fetal and adult LT-HSCs are not able to regenerate brain-resident microglia. Thus, we demonstrate that LT-HSCs retain the potential to develop into TRMΦ, but their requirement is tissue specific in the peritoneum and brain.
Collapse
Affiliation(s)
- Devon J Eddins
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA.,Division of Rheumatology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA.,Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA
| | - Astrid Kosters
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA
| | - Jeffrey Waters
- Department of Genetics, Stanford University, Stanford, CA; and
| | - Jasmine Sosa
- Department of Genetics, Stanford University, Stanford, CA; and
| | - Megan Phillips
- Department of Genetics, Stanford University, Stanford, CA; and
| | - Koshika Yadava
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | | | - Hedwich F Kuipers
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Eliver Eid Bou Ghosn
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA; .,Division of Rheumatology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA.,Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
15
|
Li Y, Magee JA. Transcriptional reprogramming in neonatal hematopoietic stem and progenitor cells. Exp Hematol 2021; 101-102:25-33. [PMID: 34303776 PMCID: PMC8557639 DOI: 10.1016/j.exphem.2021.07.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/14/2021] [Accepted: 07/19/2021] [Indexed: 02/04/2023]
Abstract
Hematopoietic stem cells (HSCs) and lineage-committed hematopoietic progenitor cells (HPCs) undergo profound shifts in gene expression during the neonatal and juvenile stages of life. Temporal changes in HSC/HPC gene expression underlie concomitant changes in self-renewal capacity, lineage biases, and hematopoietic output. Moreover, they can modify disease phenotypes. For example, childhood leukemias have distinct driver mutation profiles relative to adult leukemias, and they may arise from distinct cells of origin. The putative relationship between neonatal HSC/HPC ontogeny and childhood blood disorders highlights the importance of understanding how, at a mechanistic level, HSCs transition from fetal to adult transcriptional states. In this perspective piece, we summarize recent work indicating that the transition is uncoordinated and imprecisely timed. We discuss implications of these findings, including mechanisms that might enable neonatal HSCs and HPCs to acquire adultlike properties over a drawn-out period, in lieu of precise gene regulatory networks. The transition from fetal to adult transcriptional programs coincides with a pulse of type I interferon signaling that activates many genes associated with the adultlike state. This pulse may sensitize HSCs/HPCs to mutations that drive leukemogenesis shortly after birth. If we can understand how developmental switches modulate HSC and HPC fate after birth-both under normal circumstances and in the setting of disease-causing mutations-we can potentially reprogram these switches to treat or prevent childhood leukemias.
Collapse
|
16
|
Mack R, Zhang L, Breslin Sj P, Zhang J. The Fetal-to-Adult Hematopoietic Stem Cell Transition and its Role in Childhood Hematopoietic Malignancies. Stem Cell Rev Rep 2021; 17:2059-2080. [PMID: 34424480 DOI: 10.1007/s12015-021-10230-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 01/07/2023]
Abstract
As with most organ systems that undergo continuous generation and maturation during the transition from fetal to adult life, the hematopoietic and immune systems also experience dynamic changes. Such changes lead to many unique features in blood cell function and immune responses in early childhood. The blood cells and immune cells in neonates are a mixture of fetal and adult origin due to the co-existence of both fetal and adult types of hematopoietic stem cells (HSCs) and progenitor cells (HPCs). Fetal blood and immune cells gradually diminish during maturation of the infant and are almost completely replaced by adult types of cells by 3 to 4 weeks after birth in mice. Such features in early childhood are associated with unique features of hematopoietic and immune diseases, such as leukemia, at these developmental stages. Therefore, understanding the cellular and molecular mechanisms by which hematopoietic and immune changes occur throughout ontogeny will provide useful information for the study and treatment of pediatric blood and immune diseases. In this review, we summarize the most recent studies on hematopoietic initiation during early embryonic development, the expansion of both fetal and adult types of HSCs and HPCs in the fetal liver and fetal bone marrow stages, and the shift from fetal to adult hematopoiesis/immunity during neonatal/infant development. We also discuss the contributions of fetal types of HSCs/HPCs to childhood leukemias.
Collapse
Affiliation(s)
- Ryan Mack
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Lei Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA
| | - Peter Breslin Sj
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA.,Departments of Molecular/Cellular Physiology and Biology, Loyola University Medical Center and Loyola University Chicago, Chicago, IL, 60660, USA
| | - Jiwang Zhang
- Department of Cancer Biology, Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL, 60153, USA.
| |
Collapse
|
17
|
Pattarabanjird T, Li C, McNamara C. B Cells in Atherosclerosis: Mechanisms and Potential Clinical Applications. ACTA ACUST UNITED AC 2021; 6:546-563. [PMID: 34222726 PMCID: PMC8246059 DOI: 10.1016/j.jacbts.2021.01.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 12/17/2022]
Abstract
B cells regulate atherosclerotic plaque formation through production of antibodies and cytokines, and effects are subset specific (B1 and B2). Putative human atheroprotective B1 cells function similarly to murine B1 in their spontaneous IgM antibody production. However, marker strategies in identifying human and murine B1 are different. IgM antibody to oxidation specific epitopes produced by B1 cells associate with human coronary artery disease. Neoantigen immunization may be a promising strategy for atherosclerosis vaccine development, but further study to determine relevant antigens still need to be done. B-cell–targeted therapies, used in treating autoimmune diseases as well as lymphoid cancers, might have potential applications in treating cardiovascular diseases. Short- and long-term cardiovascular effects of these agents need to be assessed.
Because atherosclerotic cardiovascular disease is a leading cause of death worldwide, understanding inflammatory processes underpinning its pathology is critical. B cells have been implicated as a key immune cell type in regulating atherosclerosis. B-cell effects, mediated by antibodies and cytokines, are subset specific. In this review, we focus on elaborating mechanisms underlying subtype-specific roles of B cells in atherosclerosis and discuss available human data implicating B cells in atherosclerosis. We further discuss potential B cell–linked therapeutic approaches, including immunization and B cell–targeted biologics. Given recent evidence strongly supporting a role for B cells in human atherosclerosis and the expansion of immunomodulatory agents that affect B-cell biology in clinical use and clinical trials for other disorders, it is important that the cardiovascular field be cognizant of potential beneficial or untoward effects of modulating B-cell activity on atherosclerosis.
Collapse
Key Words
- APRIL, A proliferation−inducing ligand
- ApoE, apolipoprotein E
- B-cell
- BAFF, B-cell–activating factor
- BAFFR, B-cell–activating factor receptor
- BCMA, B-cell maturation antigen
- BCR, B-cell receptor
- Breg, regulatory B cell
- CAD, coronary artery disease
- CTLA4, cytotoxic T-lymphocyte–associated protein 4
- CVD, cardiovascular disease
- CXCR4, C-X-C motif chemokine receptor 4
- GC, germinal center
- GITR, glucocorticoid-induced tumor necrosis factor receptor–related protein
- GITRL, glucocorticoid-induced tumor necrosis factor receptor–related protein ligand
- GM-CSF, granulocyte-macrophage colony–stimulating factor
- ICI, immune checkpoint inhibitor
- IFN, interferon
- IL, interleukin
- IVUS, intravascular ultrasound
- LDL, low-density lipoprotein
- LDLR, low-density lipoprotein receptor
- MDA-LDL, malondialdehyde-modified low-density lipoprotein
- MI, myocardial infarction
- OSE, oxidation-specific epitope
- OxLDL, oxidized low-density lipoprotein
- PC, phosphorylcholine
- PD-1, programmed cell death protein 1
- PD-L2, programmed death ligand 2
- PDL1, programmed death ligand 1
- RA, rheumatoid arthritis
- SLE, systemic lupus erythematosus
- TACI, transmembrane activator and CAML interactor
- TNF, tumor necrosis factor
- Treg, regulatory T cell
- atherosclerosis
- immunoglobulins
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Tanyaporn Pattarabanjird
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Cynthia Li
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Coleen McNamara
- Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.,Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
18
|
Sustained fetal hematopoiesis causes juvenile death from leukemia: evidence from a dual-age-specific mouse model. Blood Adv 2021; 4:3728-3740. [PMID: 32777070 DOI: 10.1182/bloodadvances.2020002326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/16/2020] [Indexed: 11/20/2022] Open
Abstract
It is not clear whether disrupted age-specific hematopoiesis contributes to the complex manifestations in leukemia patients who carry identical mutations, particularly in pediatric and adult patients with similar clinical characteristics. By studying a dual-age-specific mouse model, we demonstrate that (1) loss of Pten during the fetal-to-adult hematopoiesis switch (hematopoiesis switch) causes sustained fetal hematopoiesis, resulting in death in juvenile leukemia; (2) myeloid-biased hematopoiesis in juvenile mice is associated with the sustained fetal properties of hematopoietic stem cells (HSCs); (3) the age specificity of juvenile myelomonocytic leukemia depends on the copy number of Pten and Nf1; (4) single-allelic Pten deletion during the hematopoiesis switch causes constitutive activation of MAPK in juvenile mice with Nf1 loss of heterozygosity (LOH); and (5) Nf1 LOH causes monocytosis in juvenile mice with Pten haploinsufficiency but does not cause lethality until adulthood. Our data suggest that 1 copy of Pten is sufficient to maintain an intact negative-feedback loop of the Akt pathway and HSC function in reconstitution, despite MAPK being constitutively activated in juvenile Pten+/ΔNf1LOH mice. However, 2 copies of Pten are required to maintain the integrity of the MAPK pathway in juvenile mice with Nf1 haploinsufficiency. Our data indicate that previous investigations of Pten function in wild-type mice may not reflect the impact of Pten loss in mice with Nf1 mutations or other genetic defects. We provide a proof of concept that disassociated age-specific hematopoiesis contributes to leukemogenesis and pediatric demise.
Collapse
|
19
|
Sakai H, Tanaka Y, Tanaka A, Ohdan H. TLR-MyD88 signaling blockades inhibit refractory B-1b cell immune responses to transplant-related glycan antigens. Am J Transplant 2021; 21:1427-1439. [PMID: 32865877 DOI: 10.1111/ajt.16288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/26/2020] [Accepted: 08/17/2020] [Indexed: 01/25/2023]
Abstract
Refractory B cell responses to T cell-independent (TI) carbohydrate antigens (Ags) are critical drivers of rejection reactions to ABO-incompatible allogeneic grafts and xenogeneic grafts from other species. To explore the biological significance of crosstalk between Toll-like receptors (TLRs) and B cell receptors (BCRs) in the TI B cell immunity, we here used MyD88-, TRIF-, and α-galactosyltransferase-deficient mice to study B cell phenotypes and functional properties during TI transplant-related glycan Ag exposure. BCR stimulation alone induced differentiation into CD5high (B-1a) cells, which were highly sensitive to a calcineurin inhibitor (CNI), while co-stimulation of TLRs and BCRs induced differentiation into CD5dim (B-1b) cells in MyD88-dependent and CNI-resistant manner. MyD88-dependent TLR stimulation in B-1b cells enhanced downstream factors in the BCR-calcineurin pathway, including a nuclear factor of activated T cells, cytoplasmic 1 (NFATc1). TLR inhibitor together with CNI abrogated refractory B-1b cell immune responses against the ABO-blood group Ags, while blocking both BCRs and TLR-MyD88 by using Bruton's tyrosine kinase inhibitor and histone deacetylase inhibitor abrogated refractory B-1b cell immune responses against Gal-glycan Ags. Thus, this study provides a rationale for a novel therapeutic approach to overcome refractory transplant-related anti-glycan Ab production by blocking both BCR and TLR-MyD88 signals.
Collapse
Affiliation(s)
- Hiroshi Sakai
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biochemical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Department of Medicine, Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biochemical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Asuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biochemical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Applied Life Sciences, Institute of Biochemical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
20
|
Jaufmann J, Tümen L, Beer-Hammer S. SLy2-overexpression impairs B-cell development in the bone marrow and the IgG response towards pneumococcal conjugate-vaccine. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:533-546. [PMID: 33592135 PMCID: PMC8127564 DOI: 10.1002/iid3.413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/08/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022]
Abstract
Background Infections with Streptococcus pneumoniae can cause severe diseases in humans including pneumonia. Although guidelines for vaccination have been established, S. pneumoniae is still responsible for a serious burden of disease around the globe. Currently, two pneumococcal immunizations are available, namely the pure polysaccharide vaccine Pneumovax23 (P23) and the conjugate‐vaccine Prevenar13 (PCV13). We recently reported impaired thymus‐independent antibody responses towards P23 in mice overexpressing the immunoinhibitory adapter SLy2. The purpose of this study was to evaluate adaptive B‐cell responses towards the thymus‐dependent vaccine PCV13 in SLy2‐overexpressing mice and to study their survival rate during pneumococcal lung infection. Moreover, we investigated B‐cell developmental stages within the bone marrow (BM) in the context of excessive SLy2‐expression. Methods B‐cell subsets and their surface immune globulins were investigated by flow cytometry. For class‐switch assays, isolated splenic B cells were stimulated in vitro with lipopolysaccharide and interleukin‐4 and antibody secretion was quantified via LEGENDplex. To study PCV13‐specific responses, mice were immunized and serum antibody titers (immunoglobulin M, immunoglobulins IgG1, IgG2, and IgG3) were examined by enzyme‐linked immunosorbent assay. Survival rates of mice were assessed within 7 days upon intranasal challenge with S. pneumoniae. Results Our data demonstrate impaired IgG1 and IgG3 antibody responses towards the pneumococcal conjugate‐vaccine PCV13 in SLy2‐overexpressing mice. This was accompanied by reduced frequencies and numbers of BM‐resident plasmablasts. In addition, we found drastically reduced counts of B‐cell precursors in the BM of SLy2‐Tg mice. The survival rate upon intranasal challenge with S. pneumoniae was mostly comparable between the genotypes. Conclusion Our findings demonstrate an important role of the adapter protein SLy2 in the context of adaptive antibody responses against pneumococcal conjugate‐vaccine. Interestingly, deficits in humoral immunity seemed to be compensated by cellular immune effectors upon bacterial challenge. Our study further shows a novel relevance of SLy2 for plasmablasts and B‐cell progenitors in the BM.
Collapse
Affiliation(s)
- Jennifer Jaufmann
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Leyla Tümen
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
21
|
Li Y, Kong W, Yang W, Patel RM, Casey EB, Okeyo-Owuor T, White JM, Porter SN, Morris SA, Magee JA. Single-Cell Analysis of Neonatal HSC Ontogeny Reveals Gradual and Uncoordinated Transcriptional Reprogramming that Begins before Birth. Cell Stem Cell 2020; 27:732-747.e7. [PMID: 32822583 PMCID: PMC7655695 DOI: 10.1016/j.stem.2020.08.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 06/21/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022]
Abstract
Fetal and adult hematopoietic stem cells (HSCs) have distinct proliferation rates, lineage biases, gene expression profiles, and gene dependencies. Although these differences are widely recognized, it is not clear how the transition from fetal to adult identity is coordinated. Here we show that murine HSCs and committed hematopoietic progenitor cells (HPCs) undergo a gradual, rather than precipitous, transition from fetal to adult transcriptional states. The transition begins prior to birth and is punctuated by a late prenatal spike in type I interferon signaling that promotes perinatal HPC expansion and sensitizes progenitors to the leukemogenic FLT3ITD mutation. Most other changes in gene expression and enhancer activation are imprecisely timed and poorly coordinated. Thus, heterochronic enhancer elements, and their associated transcripts, are activated independently of one another rather than as part of a robust network. This simplifies the regulatory programs that guide neonatal HSC/HPC ontogeny, but it creates heterogeneity within these populations.
Collapse
Affiliation(s)
- Yanan Li
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Wenjun Kong
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Riddhi M Patel
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Emily B Casey
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Theresa Okeyo-Owuor
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - J Michael White
- Department of Pathology and Immunobiology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Shaina N Porter
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Samantha A Morris
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| | - Jeffrey A Magee
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
22
|
Khalil MI, Gurski CJ, Dittel LJ, Neu SD, Dittel BN. Discovery and Function of B-Cell IgD Low (BD L) B Cells in Immune Tolerance. J Mol Biol 2020; 433:166584. [PMID: 32615130 DOI: 10.1016/j.jmb.2020.06.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/16/2020] [Accepted: 06/23/2020] [Indexed: 12/30/2022]
Abstract
It is now appreciated that in addition to their role in humoral immunity, B cells also exert regulatory mechanisms that lead to attenuation of inflammatory responses. The concept of B-cell regulation became well recognized when mice deficient in B cells due to genetic disruption were shown to be refractory to recovery from the signs of experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis. This seminal study spurred the search for B-cell regulatory phenotypes and mechanisms of action. Our approach was to utilize differential B-cell depletion with anti-CD20 to retain B cells whose presence were required to achieve EAE recovery. Utilizing flow cytometry, adoptive cell therapy and genetic approaches, we discovered a new B-cell subset that, upon adoptive transfer into B cell-deficient mice, was sufficient to promote EAE recovery. This B-cell subset is IgM+, but due to low/negative IgD cell surface expression, it was named B-cell IgD low (BDL). Mechanistically, we found that in the absence of BDL, the absolute cell number of CD4+Foxp3+ T regulatory cells (Treg), essential for immune tolerance, was significantly reduced. Furthermore, we found that BDL expression of glucocorticoid-induced tumor necrosis factor ligand (GITRL) was essential for induction of Treg proliferation and maintenance of their homeostasis. Thus, we have identified a new B-cell subset that is critical for immunological tolerance through interactions with Treg.
Collapse
Affiliation(s)
- Mohamed I Khalil
- Versiti Blood Research Institute, Milwaukee, WI, USA; Molecular Biology Department, National Research Centre, El-Buhouth St., Doki, Cairo, Egypt
| | - Cody J Gurski
- Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Landon J Dittel
- Department of Neurology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Savannah D Neu
- Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Bonnie N Dittel
- Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
23
|
Qorbani A, Gao G, Dwyre DM. Polyclonal CD5+/CD19+ B1a lymphocytes after allogeneic stem cell transplantation: a potential diagnostic pitfall. AUTOPSY AND CASE REPORTS 2020; 10:e2020147. [PMID: 33344271 PMCID: PMC7703255 DOI: 10.4322/acr.2020.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
In adults, B-lymphocytes comprise approximately 10% of circulating lymphocytes. The majority of peripheral B cells are B2 cells (“Mature” B-cells), which function as part of the humoral adaptive immune system. B1 cells (“Innate-like” B cells) are another sub-class of B lymphocytes, considered as innate immune cells with a characteristic phenotype (CD20+, CD27+, CD43+, CD70-, CD11b+, sIgM++, sIgD+) which can be divided into two subtypes; B1a (CD5+): spontaneously produce broadly reactive natural IgM, and B1b (CD5-): can generate T-cell independent, long-lasting IgM. There is very limited data available, indicating a correlation between allogeneic bone marrow transplantation and an increase in B1a cells. Here we present a case of a 17-year-old female with homozygous sickle cell disease (HbSS disease) who underwent hematopoietic stem cell transplant (HSCT). Approximately seven months post-transplant, she was found to have 16% immature mononuclear cells on complete blood count (CBC)-differential report. A follow-up peripheral blood flow cytometry showed that these cells were polyclonal CD5+/CD20+ B-cells, and comprised 66% of lymphocytes. Further workup and follow up failed to reveal any lymphoproliferative disorders. It is important not to misdiagnose these cells as an atypical CD5+ lymphoproliferative disorder. The presence of B1a cells has not been widely reported in non-neoplastic post-stem cell transplanted patients. This case also adds to and expands our knowledge regarding the presence of increased circulating B1a cells after stem cell transplant in a patient with no history of hematological malignancy.
Collapse
Affiliation(s)
- Amir Qorbani
- University of California, San Francisco (UCSF), UCSF Medical Center, Department of Pathology and Laboratory Medicine. San Francisco, CA, USA
| | - Guofeng Gao
- University of California, Davis (UC Davis), Department of Pathology and Laboratory Medicine. Sacramento, CA, USA
| | - Denis M Dwyre
- University of California, Davis (UC Davis), Department of Pathology and Laboratory Medicine. Sacramento, CA, USA
| |
Collapse
|
24
|
Kobayashi M, Lin Y, Mishra A, Shelly C, Gao R, Reeh CW, Wang PZ, Xi R, Liu Y, Wenzel P, Ghosn E, Liu Y, Yoshimoto M. Bmi1 Maintains the Self-Renewal Property of Innate-like B Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2020; 204:3262-3272. [PMID: 32332108 DOI: 10.4049/jimmunol.2000030] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/06/2020] [Indexed: 11/19/2022]
Abstract
The self-renewal ability is a unique property of fetal-derived innate-like B-1a lymphocytes, which survive and function without being replenished by bone marrow (BM) progenitors. However, the mechanism by which IgM-secreting mature B-1a lymphocytes self-renew is poorly understood. In this study, we showed that Bmi1 was critically involved in this process. Although Bmi1 is considered essential for lymphopoiesis, the number of mature conventional B cells was not altered when Bmi1 was deleted in the B cell lineage. In contrast, the number of peritoneal B-1a cells was significantly reduced. Peritoneal cell transfer assays revealed diminished self-renewal ability of Bmi1-deleted B-1a cells, which was restored by additional deletion of Ink4-Arf, the well-known target of Bmi1 Fetal liver cells with B cell-specific Bmi1 deletion failed to repopulate peritoneal B-1a cells, but not other B-2 lymphocytes after transplantation assays, suggesting that Bmi1 may be involved in the developmental process of B-1 progenitors to mature B-1a cells. Although Bmi1 deletion has also been shown to alter the microenvironment for hematopoietic stem cells, fat-associated lymphoid clusters, the reported niche for B-1a cells, were not impaired in Bmi1 -/- mice. RNA expression profiling suggested lysine demethylase 5B (Kdm5b) as another possible target of Bmi1, which was elevated in Bmi1-/- B-1a cells in a stress setting and might repress B-1a cell proliferation. Our work has indicated that Bmi1 plays pivotal roles in self-renewal and maintenance of fetal-derived B-1a cells.
Collapse
Affiliation(s)
- Michihiro Kobayashi
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Yang Lin
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Akansha Mishra
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Chris Shelly
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Rui Gao
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Colton W Reeh
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Paul Zhiping Wang
- Center for Computational Biology and Bioinformatics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Rongwen Xi
- National Institute of Biological Science, Beijing 102206, China
| | - Yunlong Liu
- Center for Computational Biology and Bioinformatics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Pamela Wenzel
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Eliver Ghosn
- Department of Medicine, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322; and.,Department of Pediatrics, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Yan Liu
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202;
| | - Momoko Yoshimoto
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030;
| |
Collapse
|
25
|
Honjo K, Won WJ, King RG, Ianov L, Crossman DK, Easlick JL, Shakhmatov MA, Khass M, Vale AM, Stephan RP, Li R, Davis RS. Fc Receptor-Like 6 (FCRL6) Discloses Progenitor B Cell Heterogeneity That Correlates With Pre-BCR Dependent and Independent Pathways of Natural Antibody Selection. Front Immunol 2020; 11:82. [PMID: 32117244 PMCID: PMC7033751 DOI: 10.3389/fimmu.2020.00082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/13/2020] [Indexed: 11/24/2022] Open
Abstract
B-1a cells produce "natural" antibodies (Abs) to neutralize pathogens and clear neo self-antigens, but the fundamental selection mechanisms that shape their polyreactive repertoires are poorly understood. Here, we identified a B cell progenitor subset defined by Fc receptor-like 6 (FCRL6) expression, harboring innate-like defense, migration, and differentiation properties conducive for natural Ab generation. Compared to FCRL6- pro B cells, the repressed mitotic, DNA damage repair, and signaling activity of FCRL6+ progenitors, yielded VH repertoires with biased distal Ighv segment accessibility, constrained diversity, and hydrophobic and charged CDR-H3 sequences. Beyond nascent autoreactivity, VH11 productivity, which predominates phosphatidylcholine-specific B-1a B cell receptors (BCRs), was higher for FCRL6+ cells as was pre-BCR formation, which was required for Myc induction and VH11, but not VH12, B-1a development. Thus, FCRL6 revealed unexpected heterogeneity in the developmental origins, regulation, and selection of natural Abs at the pre-BCR checkpoint with implications for autoimmunity and lymphoproliferative disorders.
Collapse
MESH Headings
- Animals
- Antibodies/immunology
- Antibodies/metabolism
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Female
- Humans
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Heavy Chains/immunology
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Phosphatidylcholines/immunology
- Phosphatidylcholines/metabolism
- Precursor Cells, B-Lymphoid/immunology
- Precursor Cells, B-Lymphoid/metabolism
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Fc/genetics
- Receptors, Fc/immunology
- Receptors, Fc/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
Collapse
Affiliation(s)
- Kazuhito Honjo
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Woong-Jai Won
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rodney G. King
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lara Ianov
- Civitan International Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - David K. Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Juliet L. Easlick
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mikhail A. Shakhmatov
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mohamed Khass
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Genetic Engineering and Biotechnology Division, National Research Center, Cairo, Egypt
| | - Andre M. Vale
- Program in Immunobiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Robert P. Stephan
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ran Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Randall S. Davis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
26
|
Wang Y, Liu J, Burrows PD, Wang JY. B Cell Development and Maturation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1254:1-22. [PMID: 32323265 DOI: 10.1007/978-981-15-3532-1_1] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Since the identification of B cells in 1965 (Cooper et al. 1965), three has been tremendous progress in our understanding of B cell development, maturation and function. A number of B cell subpopulations, including B-1, B-2 and regulatory B cells, have been identified. B-1 cells mainly originate from the fetal liver and contain B-1a and B-1b subsets. B-2 cells are derived from the bone marrow (BM) and can be further classified into follicular B (FOB) and marginal zone B (MZB) cells. Regulatory B cells (Bregs) function to suppress immune responses, primarily by production of the anti-inflammatory cytokine IL-10. B cell tolerance is established at several checkpoints, during B cell development in the BM (central tolerance) as well as during B cell maturation and activation in the periphery (peripheral tolerance). This chapter will focus on the regulation of important processes during the development and maturation of B-1 and B-2 cells.
Collapse
Affiliation(s)
- Ying Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jun Liu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Peter D Burrows
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
27
|
The ontogeny of murine B-1a cells. Int J Hematol 2019; 111:622-627. [PMID: 31802412 DOI: 10.1007/s12185-019-02787-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 10/25/2022]
Abstract
It has been over 35 years since the discovery of a special subtype of B cells in mice. These IgM+ B cells are named B-1 cells, whereas conventional B cells are referred to as B-2 cells. B-1 cells express Ly-1 (CD5) and CD11b antigen, which are usually expressed in T cells and myeloid cells, respectively, reside mainly in the peritoneal and pleural cavities, and secrete natural IgM antibodies in a T cell-independent manner. B-1 cells are further categorized into CD5+ B-1a cells and CD5- B-1b cells. B-1 cells may develop through positive selection and secrete natural antibodies, including low-affinity-binding autoantibodies. Transplantation assays have revealed that the fetal liver, not the bone marrow (BM), is a major site for the production of B-1a cells, leading to the concept of a fetal origin for B-1a cells. This review introduces how the origin of B-1a cells has been explored, and describes the current state of knowledge gained through various approaches.
Collapse
|
28
|
Kikushige Y. Pathophysiology of chronic lymphocytic leukemia and human B1 cell development. Int J Hematol 2019; 111:634-641. [PMID: 31797231 DOI: 10.1007/s12185-019-02788-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 12/22/2022]
Abstract
Chronic lymphocytic leukemia (CLL), the most frequent type of leukemia in adults, is a lymphoproliferative disease characterized by the clonal expansion of mature CD5+ B cells in peripheral blood, bone marrow, and secondary lymphoid tissues. Over the past decade, substantial advances have been made in understanding the pathogenesis of CLL, including the identification of recurrent mutations, and clarification of clonal architectures, transcriptome analyses, and the multistep leukemogenic process. The biology of CLL is now better understood. The present review focuses on recent insights into CLL leukemogenesis, emphasizing the role of genetic lesions, and the multistep process initiating from very immature hematopoietic stem cells. Finally, we also review progress in the study of human B1 B cells, the putative normal counterparts of CLL cells.
Collapse
Affiliation(s)
- Yoshikane Kikushige
- Department of Medicine and Biosystemic Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.
| |
Collapse
|
29
|
Ghosn E, Yoshimoto M, Nakauchi H, Weissman IL, Herzenberg LA. Hematopoietic stem cell-independent hematopoiesis and the origins of innate-like B lymphocytes. Development 2019; 146:146/15/dev170571. [PMID: 31371526 DOI: 10.1242/dev.170571] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The current paradigm that a single long-term hematopoietic stem cell can regenerate all components of the mammalian immune system has been challenged by recent findings in mice. These findings show that adult tissue-resident macrophages and innate-like lymphocytes develop early in fetal hematopoiesis from progenitors that emerge prior to, and apparently independently of, conventional long-term hematopoietic stem cells. Here, we discuss these recent findings, which show that an early and distinct wave of hematopoiesis occurs for all major hematopoietic lineages. These data provide evidence that fetal hematopoietic progenitors not derived from the bona fide long-term hematopoietic stem cells give rise to tissue-resident immune cells that persist throughout adulthood. We also discuss recent insights into B lymphocyte development and attempt to synthesize seemingly contradictory recent findings on the origins of innate-like B-1a lymphocytes during fetal hematopoiesis.
Collapse
Affiliation(s)
- Eliver Ghosn
- Departments of Medicine and Pediatrics, Lowance Center for Human Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Momoko Yoshimoto
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hiromitsu Nakauchi
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Leonore A Herzenberg
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
30
|
Kobayashi M, Tarnawsky SP, Wei H, Mishra A, Azevedo Portilho N, Wenzel P, Davis B, Wu J, Hadland B, Yoshimoto M. Hemogenic Endothelial Cells Can Transition to Hematopoietic Stem Cells through a B-1 Lymphocyte-Biased State during Maturation in the Mouse Embryo. Stem Cell Reports 2019; 13:21-30. [PMID: 31231025 PMCID: PMC6626887 DOI: 10.1016/j.stemcr.2019.05.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 11/27/2022] Open
Abstract
Precursors of hematopoietic stem cells (pre-HSCs) have been identified as intermediate precursors during the maturation process from hemogenic endothelial cells to HSCs in the aorta-gonad-mesonephros (AGM) region of the mouse embryo at embryonic day 10.5. Although pre-HSCs acquire an efficient adult-repopulating ability after ex vivo co-culture, their native hematopoietic capacity remains unknown. Here, we employed direct transplantation assays of CD45-VE-cadherin(VC)+KIT+(V+K+) cells (containing pre-HSCs) into immunodeficient neonatal mice that permit engraftment of embryonic hematopoietic precursors. We found that freshly isolated V+K+ cells exhibited significantly greater B-1 lymphocyte-biased repopulating capacity than multilineage repopulating capacity. Additionally, B cell colony-forming assays demonstrated the predominant B-1 progenitor colony-forming ability of these cells; however, increased B-2 progenitor colony-forming ability emerged after co-culture with Akt-expressing AGM endothelial cells, conditions that support pre-HSC maturation into HSCs. Our studies revealed an unexpected B-1 lymphocyte bias of the V+K+ population and acquisition of B-2 potential during commitment to the HSC fate.
Collapse
Affiliation(s)
- Michihiro Kobayashi
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Stefan P Tarnawsky
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Haichao Wei
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Akansha Mishra
- Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nathalia Azevedo Portilho
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Pamela Wenzel
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brian Davis
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jiaqian Wu
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brandon Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Washington School of Medicine, Seattle, WA 98105, USA
| | - Momoko Yoshimoto
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
31
|
Lin Y, Kobayashi M, Azevedo Portilho N, Mishra A, Gao H, Liu Y, Wenzel P, Davis B, Yoder MC, Yoshimoto M. Long-Term Engraftment of ESC-Derived B-1 Progenitor Cells Supports HSC-Independent Lymphopoiesis. Stem Cell Reports 2019; 12:572-583. [PMID: 30745034 PMCID: PMC6409422 DOI: 10.1016/j.stemcr.2019.01.006] [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: 05/13/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 01/09/2023] Open
Abstract
It is generally considered that mouse embryonic stem cell (ESC) differentiation into blood cells in vitro recapitulates yolk sac (YS) hematopoiesis. As such, similar to YS-derived B-progenitors, we demonstrate here that ESC-derived B-progenitors differentiate into B-1 and marginal zone B cells, but not B-2 cells in immunodeficient mice after transplantation. ESC-derived B-1 cells were maintained in the recipients for more than 6 months, secreting natural IgM antibodies in vivo. Gene expression profiling displayed a close relationship between ESC- and YS-derived B-1 progenitors. Because there are no hematopoietic stem cells (HSCs) detectable in our ESC differentiation culture, successful long-term engraftment of ESC-derived functional B-1 cells supports the presence of HSC-independent B-1 cell development. ESC-derived B-progenitors mature into B-1 cells and MZ B cells in vivo ESC-derived B-1 cells engrafted in vivo long-term and secrete natural antibodies ESC-derived B-progenitors are molecularly similar to YS-derived B-progenitors Long-term B-1 cell engraftment represents HSC-independent lymphopoiesis
Collapse
Affiliation(s)
- Yang Lin
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Michihiro Kobayashi
- Center for Stem Cell Research, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nathalia Azevedo Portilho
- Center for Stem Cell Research, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Akansha Mishra
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Pamela Wenzel
- Center for Stem Cell Research, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Brian Davis
- Center for Stem Cell Research, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mervin C Yoder
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Momoko Yoshimoto
- Center for Stem Cell Research, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Abstract
Hematopoiesis is the process by which mature blood and immune cells are produced from hematopoietic stem and progenitor cells (HSCs and HSPCs). The last several decades of research have shed light on the origin of HSCs, as well as the heterogeneous pools of fetal progenitors that contribute to lifelong hematopoiesis. The overarching concept that hematopoiesis occurs in dynamic, overlapping waves throughout development, with each wave contributing to both continuous and developmentally limited cell types, has been solidified over the years. However, recent advances in our ability to track the production of hematopoietic cells in vivo have challenged several long-held dogmas on the origin and persistence of distinct hematopoietic cell types. In this review, we highlight emerging concepts in hematopoietic development and identify unanswered questions.
Collapse
Affiliation(s)
- Taylor Cool
- Institute for the Biology of Stem Cells, Program in Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA, United States.
| |
Collapse
|
33
|
Mature IgD low/- B cells maintain tolerance by promoting regulatory T cell homeostasis. Nat Commun 2019; 10:190. [PMID: 30643147 PMCID: PMC6331566 DOI: 10.1038/s41467-018-08122-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 12/19/2018] [Indexed: 12/31/2022] Open
Abstract
A number of different B cell subsets have been shown to exhibit regulatory activity using a variety of mechanisms to attenuate inflammatory diseases. Here we show, using anti-CD20-mediated partial B cell depletion in mice, that a population of mature B cells distinguishable by IgDlow/- expression maintains tolerance by, at least in part, promoting CD4+Foxp3+ regulatory T cell homeostatic expansion via glucocorticoid-induced tumor necrosis factor receptor ligand, or GITRL. Cell surface phenotyping, transcriptome analysis and developmental study data show that B cells expressing IgD at a low level (BDL) are a novel population of mature B cells that emerge in the spleen from the transitional-2 stage paralleling the differentiation of follicular B cells. The cell surface phenotype and regulatory function of BDL are highly suggestive that they are a new B cell subset. Human splenic and peripheral blood IgDlow/- B cells also exhibit BDL regulatory activity, rendering them of therapeutic interest.
Collapse
|
34
|
Fereidan-Esfahani M, Nayfeh T, Warrington A, Howe CL, Rodriguez M. IgM Natural Autoantibodies in Physiology and the Treatment of Disease. Methods Mol Biol 2019; 1904:53-81. [PMID: 30539466 DOI: 10.1007/978-1-4939-8958-4_3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Antibodies are vital components of the adaptive immune system for the recognition and response to foreign antigens. However, some antibodies recognize self-antigens in healthy individuals. These autoreactive antibodies may modulate innate immune functions. IgM natural autoantibodies (IgM-NAAs) are a class of primarily polyreactive immunoglobulins encoded by germline V-gene segments which exhibit low affinity but broad specificity to both foreign and self-antigens. Historically, these autoantibodies were closely associated with autoimmune disease. Nevertheless, not all human autoantibodies are pathogenic and compelling evidence indicates that IgM-NAAs may exert a spectrum of effects from injurious to protective depending upon cellular and molecular context. In this chapter, we review the current state of knowledge regarding the potential physiological and therapeutic roles of IgM-NAAs in different disease conditions such as atherosclerosis, cancer, and autoimmune disease. We also describe the discovery of two reparative IgM-NAAs by our laboratory and delineate their proposed mechanisms of action in central nervous system (CNS) disease.
Collapse
Affiliation(s)
| | - Tarek Nayfeh
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Charles L Howe
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
| | | |
Collapse
|
35
|
Koga S, Hozumi K, Hirano KI, Yazawa M, Terooatea T, Minoda A, Nagasawa T, Koyasu S, Moro K. Peripheral PDGFRα +gp38 + mesenchymal cells support the differentiation of fetal liver-derived ILC2. J Exp Med 2018; 215:1609-1626. [PMID: 29728440 PMCID: PMC5987924 DOI: 10.1084/jem.20172310] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 04/05/2018] [Accepted: 04/11/2018] [Indexed: 01/01/2023] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are derived from common lymphoid progenitors (CLPs) via several specific precursors, and the transcription factors essential for ILC2 differentiation have been extensively studied. However, the external factors regulating commitment to the ILC lineage as well as the sites and stromal cells that constitute the optimal microenvironment for ILC2-specific differentiation are not fully defined. In this study, we demonstrate that three key external factors, the concentration of interleukin 7 (IL-7) and strength and duration of Notch signaling, coordinately determine the fate of CLP toward the T, B, or ILC lineage. Additionally, we identified three stages of ILC2 in the fetal mesentery that require STAT5 signals for maturation: ILC progenitors, CCR9+ ILC2 progenitors, and KLRG1- immature ILC2. We further demonstrate that ILC2 development is supported by mesenteric platelet-derived growth factor receptor α (PDGFRα)+ glycoprotein 38 (gp38)+ mesenchymal cells. Collectively, our results suggest that early differentiation of ILC2 occurs in the fetal liver via IL-7 and Notch signaling, whereas final differentiation occurs in the periphery with the aid of PDGFRα+gp38+ cells.
Collapse
Affiliation(s)
- Satoshi Koga
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Division of Immunobiology, Department of Medical Life Science, Department of Supramolecular Biology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Katsuto Hozumi
- Department of Immunology, Tokai University School of Medicine, Isehara, Japan
| | - Ken-Ichi Hirano
- Department of Immunology, Tokai University School of Medicine, Isehara, Japan
| | - Masaki Yazawa
- Department of Immunology, Tokai University School of Medicine, Isehara, Japan.,Department of Biochemistry, Tokai University, Hiratsuka, Japan
| | - Tommy Terooatea
- Epigenome Technology Exploration Unit, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Aki Minoda
- Epigenome Technology Exploration Unit, RIKEN Center for Life Science Technologies, Yokohama, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Deparment of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan .,Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Division of Immunobiology, Department of Medical Life Science, Department of Supramolecular Biology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|
36
|
Rosado MM, Aranburu A, Scarsella M, Cascioli S, Giorda E, Del Chierico F, Mortera SL, Mortari EP, Petrini S, Putignani L, Carsetti R. Spleen development is modulated by neonatal gut microbiota. Immunol Lett 2018; 199:1-15. [PMID: 29715493 DOI: 10.1016/j.imlet.2018.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/27/2018] [Accepted: 04/27/2018] [Indexed: 01/11/2023]
Abstract
The full development of the mammalian immune system occurs after birth upon exposure to non self-antigens. The gut is the first site of bacterial colonization where it is crucial to create the appropriate microenvironment able to balance effector or tolerogenic responses to external stimuli. It is a well-established fact that at mucosal sites bacteria play a key role in developing the immune system but we ignore how colonising bacteria impact the maturation of the spleen. Here we addressed this issue. Taking advantage of the fact that milk SIgA regulates bacterial colonization of the newborn intestine, we generated immunocompetent mice born either from IgA pro-efficient or IgA deficient females. Having demonstrated that SIgA in maternal milk modulates neonatal gut microbiota by promoting an increased diversity of the colonizing species we also found that immunocompetent pups, not exposed to milk SIgA, fail to properly develop the FDC network and primary follicles in the spleen compromising the response to T-dependent antigens. The presence of a less diverse microbiota with a higher representation of pathogenic species leads to a fast replenishment of the marginal zone and the IgM plasma cell compartment of the spleen as well as IgA plasma cells in the gut.
Collapse
Affiliation(s)
- M Manuela Rosado
- B Cell Physiopathology Unit, Immunology Research Area, Bambino Gesù Children Hospital, Rome, Italy.
| | - Alaitz Aranburu
- B Cell Physiopathology Unit, Immunology Research Area, Bambino Gesù Children Hospital, Rome, Italy
| | - Marco Scarsella
- B Cell Physiopathology Unit, Immunology Research Area, Bambino Gesù Children Hospital, Rome, Italy
| | - Simona Cascioli
- B Cell Physiopathology Unit, Immunology Research Area, Bambino Gesù Children Hospital, Rome, Italy
| | - Ezio Giorda
- B Cell Physiopathology Unit, Immunology Research Area, Bambino Gesù Children Hospital, Rome, Italy
| | - Federica Del Chierico
- Human Microbiome Unit, Area of Genetic and Rare Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Stefano Levi Mortera
- Human Microbiome Unit, Area of Genetic and Rare Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Eva Piano Mortari
- B Cell Physiopathology Unit, Immunology Research Area, Bambino Gesù Children Hospital, Rome, Italy
| | - Stefania Petrini
- Confocal Microscopy Core Facility, Bambino Gesù Children's Hospital, Rome, Italy
| | - Lorenza Putignani
- Human Microbiome Unit, Area of Genetic and Rare Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | - Rita Carsetti
- B Cell Physiopathology Unit, Immunology Research Area, Bambino Gesù Children Hospital, Rome, Italy
| |
Collapse
|
37
|
Prohaska TA, Que X, Diehl CJ, Hendrikx S, Chang MW, Jepsen K, Glass CK, Benner C, Witztum JL. Massively Parallel Sequencing of Peritoneal and Splenic B Cell Repertoires Highlights Unique Properties of B-1 Cell Antibodies. THE JOURNAL OF IMMUNOLOGY 2018; 200:1702-1717. [PMID: 29378911 PMCID: PMC5821571 DOI: 10.4049/jimmunol.1700568] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 01/03/2018] [Indexed: 12/26/2022]
Abstract
B-1 cells are a unique subset of B cells that are positively selected for expressing autoreactive BCRs. We isolated RNA from peritoneal (B-1a, B-1b, B-2) and splenic (B-1a, marginal zone, follicular) B cells from C57BL/6 mice and used 5'-RACE to amplify the IgH V region using massively parallel sequencing. By analyzing 379,000 functional transcripts, we demonstrate that B-1a cells use a distinct and restricted repertoire. All B-1 cell subsets, especially peritoneal B-1a cells, had a high proportion of sequences without N additions, suggesting predominantly prenatal development. Their transcripts differed markedly and uniquely contained VH11 and VH12 genes, which were rearranged only with a restricted selection of D and J genes, unlike other V genes. Compared to peritoneal B-1a, the peritoneal B-1b repertoire was larger, had little overlap with B-1a, and most sequences contained N additions. Similarly, the splenic B-1a repertoire differed from peritoneal B-1a sequences, having more unique sequences and more frequent N additions, suggesting influx of B-1a cells into the spleen from nonperitoneal sites. Two CDR3s, previously described as Abs to bromelain-treated RBCs, comprised 43% of peritoneal B-1a sequences. We show that a single-chain variable fragment designed after the most prevalent B-1a sequence bound oxidation-specific epitopes such as the phosphocholine of oxidized phospholipids. In summary, we provide the IgH V region library of six murine B cell subsets, including, to our knowledge for the first time, a comparison between B-1a and B-1b cells, and we highlight qualities of B-1 cell Abs that indicate unique selection processes.
Collapse
Affiliation(s)
- Thomas A Prohaska
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Xuchu Que
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Cody J Diehl
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Sabrina Hendrikx
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Max W Chang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Kristen Jepsen
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA 92093; and
| | - Christopher K Glass
- Department of Medicine, University of California San Diego, La Jolla, CA 92093.,Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093
| | - Christopher Benner
- Department of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Joseph L Witztum
- Department of Medicine, University of California San Diego, La Jolla, CA 92093;
| |
Collapse
|
38
|
Hadland B, Yoshimoto M. Many layers of embryonic hematopoiesis: new insights into B-cell ontogeny and the origin of hematopoietic stem cells. Exp Hematol 2017; 60:1-9. [PMID: 29287940 DOI: 10.1016/j.exphem.2017.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 12/22/2022]
Abstract
In adult hematopoiesis, the hematopoietic stem cell (HSC) sits at the top of a hierarchy of hematopoietic progenitors responsible for generating the diverse repertoire of blood and immune cells. During embryonic development, however, the initial waves of hematopoiesis provide the first functioning blood cells of the developing embryo, such as primitive erythrocytes arising in the yolk sac, independently of HSCs. In the field of developmental immunology, it has been recognized that some components of the immune system, such as B-1a lymphocytes, are uniquely produced during the embryonic and neonatal period, suggesting a "layered" development of immunity. Several recent studies have shed new light on the developmental origin of the layered immune system, suggesting complex and sometimes multiple contributions to unique populations of innate-like immune cells from both fetal HSCs and earlier HSC-independent progenitors. In this review, we will attempt to synthesize these studies to provide an integrated model of developmental hematopoiesis and layered immunity that may offer new insights into the origin of HSCs.
Collapse
Affiliation(s)
- Brandon Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Washington School of Medicine, Seattle, WA, USA.
| | - Momoko Yoshimoto
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
39
|
Savage HP, Yenson VM, Sawhney SS, Mousseau BJ, Lund FE, Baumgarth N. Blimp-1-dependent and -independent natural antibody production by B-1 and B-1-derived plasma cells. J Exp Med 2017; 214:2777-2794. [PMID: 28698287 PMCID: PMC5584113 DOI: 10.1084/jem.20161122] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 04/27/2017] [Accepted: 06/08/2017] [Indexed: 11/04/2022] Open
Abstract
Natural antibodies contribute to tissue homeostasis and protect against infections. They are secreted constitutively without external antigenic stimulation. The differentiation state and regulatory pathways that enable continuous natural antibody production by B-1 cells, the main cellular source in mice, remain incompletely understood. Here we demonstrate that natural IgM-secreting B-1 cells in the spleen and bone marrow are heterogeneous, consisting of (a) terminally differentiated B-1-derived plasma cells expressing the transcriptional regulator of differentiation, Blimp-1, (b) Blimp-1+, and (c) Blimp-1neg phenotypic B-1 cells. Blimp-1neg IgM-secreting B-1 cells are not simply intermediates of cellular differentiation. Instead, they secrete similar amounts of IgM in wild-type and Blimp-1-deficient (PRDM-1ΔEx1A) mice. Blimp-1neg B-1 cells are also a major source of IgG3. Consequently, deletion of Blimp-1 changes neither serum IgG3 levels nor the amount of IgG3 secreted per cell. Thus, the pool of natural antibody-secreting B-1 cells is heterogeneous and contains a distinct subset of cells that do not use Blimp-1 for initiation or maximal antibody secretion.
Collapse
Affiliation(s)
- Hannah P Savage
- Graduate Group in Immunology, University of California, Davis, Davis, CA.,Center for Comparative Medicine, University of California, Davis, Davis, CA
| | - Vanessa M Yenson
- Center for Comparative Medicine, University of California, Davis, Davis, CA
| | - Sanjam S Sawhney
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA
| | - Betty J Mousseau
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - Frances E Lund
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL
| | - Nicole Baumgarth
- Graduate Group in Immunology, University of California, Davis, Davis, CA .,Center for Comparative Medicine, University of California, Davis, Davis, CA.,Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA
| |
Collapse
|
40
|
Hadland BK, Varnum-Finney B, Mandal PK, Rossi DJ, Poulos MG, Butler JM, Rafii S, Yoder MC, Yoshimoto M, Bernstein ID. A Common Origin for B-1a and B-2 Lymphocytes in Clonal Pre- Hematopoietic Stem Cells. Stem Cell Reports 2017; 8:1563-1572. [PMID: 28479303 PMCID: PMC5469918 DOI: 10.1016/j.stemcr.2017.04.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022] Open
Abstract
Recent evidence points to the embryonic emergence of some tissue-resident innate immune cells, such as B-1a lymphocytes, prior to and independently of hematopoietic stem cells (HSCs). However, whether the full hematopoietic repertoire of embryonic HSCs initially includes these unique lineages of innate immune cells has been difficult to assess due to lack of clonal assays that identify and assess HSC precursor (pre-HSC) potential. Here, by combining index sorting of single embryonic hemogenic precursors with in vitro HSC maturation and transplantation assays, we analyze emerging pre-HSCs at the single-cell level, revealing their unique stage-specific properties and clonal lineage potential. Remarkably, clonal pre-HSCs detected between E9.5 and E11.5 contribute to the complete B cell repertoire, including B-1a lymphocytes, revealing a previously unappreciated common precursor for all B cell lineages at the pre-HSC stage and a second embryonic origin for B-1a lymphocytes. Index sorting and stromal co-culture identifies clonal embryonic pre-HSCs Clonal pre-HSCs have both B-1a and B-2 lymphocyte potential Clonal pre-HSCs express distinctive levels of Delta-like-4
Collapse
Affiliation(s)
- Brandon K Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D2-373, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, USA.
| | - Barbara Varnum-Finney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D2-373, Seattle, WA 98109, USA
| | - Pankaj K Mandal
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Division of Hematology/Oncology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02116, USA
| | - Derrick J Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Division of Hematology/Oncology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02116, USA
| | - Michael G Poulos
- Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10021, USA; Department of Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Jason M Butler
- Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10021, USA; Department of Surgery, Weill Cornell Medical College, New York, NY 10021, USA
| | - Shahin Rafii
- Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medical College, New York, NY 10021, USA
| | - Mervin C Yoder
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Momoko Yoshimoto
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Irwin D Bernstein
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue N, D2-373, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, USA
| |
Collapse
|
41
|
To B1a or not to B1a: do hematopoietic stem cells contribute to tissue-resident immune cells? Blood 2016; 128:2765-2769. [PMID: 27799163 DOI: 10.1182/blood-2016-10-697813] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/25/2016] [Indexed: 12/23/2022] Open
Abstract
Hematopoietic stem cells (HSCs) have long been considered the continuous source of all hematopoietic cells for the life of an individual. Recent findings have questioned multiple aspects of this view, including the ability of lifelong HSCs to contribute to tissue-resident immune cells. Here we discuss the most recent findings on the source of B1a cells, innatelike lymphocytes that primarily reside in serous cavities. Powerful experimental approaches including bar coding, single cell transplantation, in vivo lineage tracing, and HSC-specific pulse-chase labeling have provided novel insights on B1a-cell generation during ontogeny. We evaluate the evidence for fetal vs adult B1a-cell production capacity and the identity of putative cells of origin. Integrating these most recent findings with previous work, we propose a working model that encapsulates our current understanding of waves of immune development.
Collapse
|
42
|
Fetal Lymphoid Progenitors Become Restricted to B-1 Fates Coincident with IL-7Rα Expression. PLoS One 2016; 11:e0165676. [PMID: 27792746 PMCID: PMC5085042 DOI: 10.1371/journal.pone.0165676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 10/14/2016] [Indexed: 11/19/2022] Open
Abstract
B-1 cells represent a sub-fraction of B lymphocytes that participate in T cell-independent antibody production and contribute to innate immunity. While the production of B-1 cells is favored during the fetal waves of lymphopoiesis, it has been unclear when and how that differentiation option is specified. To clarify this, lymphoid and hematopoietic progenitors of fetal liver (FL) and adult bone marrow (ABM) were examined for the B cell differentiation potential. Mouse common lymphoid progenitors (CLPs) and more primitive KSL fraction of FL and ABM were transferred to SCID mice and donor-derived B cell subsets were analyzed 4 weeks later. CLPs were also cultured on ST2 stromal cells for 6 days prior to transplantation. While Lin- IL-7Rα+ CLPs from ABM differentiated to B-1, B-2 and marginal zone B (MZB) cells, equivalent cells from d15 FL differentiated mostly to B-1a cells. We found that fetal CLPs had less ability to colonize the bone marrow than adult CLPs. However, the fetal/adult difference was already present when progenitors were cultured in an identical condition before transplantation. More primitive KSL fraction of FL could generate the same broad spectrum of B cells typical of adults, including splenic MZB cells. In conclusion, we argue that FL and ABM-CLPs are intrinsically different regarding B-1/B-2 fates and the difference is acquired just before or coincident with the acquisition of IL-7Rα expression.
Collapse
|
43
|
Quách TD, Hopkins TJ, Holodick NE, Vuyyuru R, Manser T, Bayer RL, Rothstein TL. Human B-1 and B-2 B Cells Develop from Lin-CD34+CD38lo Stem Cells. THE JOURNAL OF IMMUNOLOGY 2016; 197:3950-3958. [PMID: 27815443 DOI: 10.4049/jimmunol.1600630] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 09/14/2016] [Indexed: 12/24/2022]
Abstract
The B-1 B cell population is an important bridge between innate and adaptive immunity primarily because B-1 cells produce natural Ab. Murine B-1 and B-2 cells arise from distinct progenitors; however, in humans, in part because it has been difficult to discriminate between them phenotypically, efforts to pinpoint the developmental origins of human B-1 and B-2 cells have lagged. To characterize progenitors of human B-1 and B-2 cells, we separated cord blood and bone marrow Lin-CD34+ hematopoietic stem cells into Lin-CD34+CD38lo and Lin-CD34+CD38hi populations. We found that transplanted Lin-CD34+CD38lo cells, but not Lin-CD34+CD38hi cells, generated a CD19+ B cell population after transfer into immunodeficient NOD.Cg-Prkdcscid Il2rgtm1wjl/SxJ neonates. The emergent CD19+ B cell population was found in spleen, bone marrow, and peritoneal cavity of humanized mice and included distinct populations displaying the B-1 or the B-2 cell phenotype. Engrafted splenic B-1 cells exhibited a mature phenotype, as evidenced by low-to-intermediate expression levels of CD24 and CD38. The engrafted B-1 cell population expressed a VH-DH-JH composition similar to cord blood B-1 cells, including frequent use of VH4-34 (8 versus 10%, respectively). Among patients with hematologic malignancies who underwent hematopoietic stem cell transplantation, B-1 cells were found in the circulation as early as 8 wk posttransplantation. Altogether, our data demonstrate that human B-1 and B-2 cells develop from a Lin-CD34+CD38lo stem cell population, and engrafted B-1 cells in humanized mice exhibit an Ig-usage pattern comparable to B-1 cells in cord blood.
Collapse
Affiliation(s)
- Tâm D Quách
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Thomas J Hopkins
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Nichol E Holodick
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Raja Vuyyuru
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Tim Manser
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Ruthee-Lu Bayer
- Monter Cancer Center, North Shore University Hospital, Northwell Health, Lake Success, NY 11042; and
| | - Thomas L Rothstein
- Center for Oncology and Cell Biology, The Feinstein Institute for Medical Research, Manhasset, NY 11030; .,Hofstra-Northwell Health School of Medicine, Hempstead, NY 11549
| |
Collapse
|
44
|
Beaudin AE, Boyer SW, Perez-Cunningham J, Hernandez GE, Derderian SC, Jujjavarapu C, Aaserude E, MacKenzie T, Forsberg EC. A Transient Developmental Hematopoietic Stem Cell Gives Rise to Innate-like B and T Cells. Cell Stem Cell 2016; 19:768-783. [PMID: 27666010 DOI: 10.1016/j.stem.2016.08.013] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 06/01/2016] [Accepted: 08/15/2016] [Indexed: 12/24/2022]
Abstract
The generation of distinct hematopoietic cell types, including tissue-resident immune cells, distinguishes fetal from adult hematopoiesis. However, the mechanisms underlying differential cell production to generate a layered immune system during hematopoietic development are unclear. Using an irreversible lineage-tracing model, we identify a definitive hematopoietic stem cell (HSC) that supports long-term multilineage reconstitution upon transplantation into adult recipients but does not persist into adulthood in situ. These HSCs are fully multipotent, yet they display both higher lymphoid cell production and greater capacity to generate innate-like B and T lymphocytes as compared to coexisting fetal HSCs and adult HSCs. Thus, these developmentally restricted HSCs (drHSCs) define the origin and generation of early lymphoid cells that play essential roles in establishing self-recognition and tolerance, with important implications for understanding autoimmune disease, allergy, and rejection of transplanted organs.
Collapse
Affiliation(s)
- Anna E Beaudin
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Scott W Boyer
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jessica Perez-Cunningham
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Gloria E Hernandez
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - S Christopher Derderian
- Eli and Edythe Broad Center of Regeneration Medicine and Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chethan Jujjavarapu
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Eric Aaserude
- Institute for the Biology of Stem Cells, Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tippi MacKenzie
- Eli and Edythe Broad Center of Regeneration Medicine and Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - 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
|
45
|
Montecino-Rodriguez E, Fice M, Casero D, Berent-Maoz B, Barber CL, Dorshkind K. Distinct Genetic Networks Orchestrate the Emergence of Specific Waves of Fetal and Adult B-1 and B-2 Development. Immunity 2016; 45:527-539. [PMID: 27566938 DOI: 10.1016/j.immuni.2016.07.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/13/2016] [Accepted: 06/08/2016] [Indexed: 10/21/2022]
Abstract
B cell development is often depicted as a linear process initiating in the fetus and continuing postnatally. Using a PU.1 hypomorphic mouse model, we found that B-1 and B-2 lymphopoiesis occurred in distinct fetal and adult waves differentially dependent on the Sfpi1 14 kB upstream regulatory element. The initial wave of fetal B-1 development was absent in PU.1 hypomorphic mice, while subsequent fetal and adult waves emerged. In contrast, B-2 lymphopoiesis occurred in distinct fetal and adult waves. Whole-transcriptome profiling of fetal and adult B cell progenitors supported the existence of three waves of B-1 and two waves of B-2 development and revealed that the network of transcription factors governing B lineage specification and commitment was highly divergent between B-1 and B-2 progenitors. These findings support the view that the B-1 and B-2 lineages are distinct and provide a genetic basis for layering of immune system development.
Collapse
Affiliation(s)
- Encarnacion Montecino-Rodriguez
- Department of Pathology and Laboratory Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michael Fice
- Department of Pathology and Laboratory Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - David Casero
- Department of Pathology and Laboratory Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Beata Berent-Maoz
- Department of Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chad L Barber
- Department of Biology, California Lutheran University, Thousand Oaks, CA 91360, USA
| | - Kenneth Dorshkind
- Department of Pathology and Laboratory Medicine, Division of Hematology/Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
46
|
Cellular Barcoding Links B-1a B Cell Potential to a Fetal Hematopoietic Stem Cell State at the Single-Cell Level. Immunity 2016; 45:346-57. [DOI: 10.1016/j.immuni.2016.07.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/30/2016] [Accepted: 04/27/2016] [Indexed: 11/18/2022]
|
47
|
Frenkel D, Zhang F, Guirnalda P, Haynes C, Bockstal V, Radwanska M, Magez S, Black SJ. Trypanosoma brucei Co-opts NK Cells to Kill Splenic B2 B Cells. PLoS Pathog 2016; 12:e1005733. [PMID: 27403737 PMCID: PMC4942092 DOI: 10.1371/journal.ppat.1005733] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/08/2016] [Indexed: 11/19/2022] Open
Abstract
After infection with T. brucei AnTat 1.1, C57BL/6 mice lost splenic B2 B cells and lymphoid follicles, developed poor parasite-specific antibody responses, lost weight, became anemic and died with fulminating parasitemia within 35 days. In contrast, infected C57BL/6 mice lacking the cytotoxic granule pore-forming protein perforin (Prf1-/-) retained splenic B2 B cells and lymphoid follicles, developed high-titer antibody responses against many trypanosome polypeptides, rapidly suppressed parasitemia and did not develop anemia or lose weight for at least 60 days. Several lines of evidence show that T. brucei infection-induced splenic B cell depletion results from natural killer (NK) cell-mediated cytotoxicity: i) B2 B cells were depleted from the spleens of infected intact, T cell deficient (TCR-/-) and FcγRIIIa deficient (CD16-/-) C57BL/6 mice excluding a requirement for T cells, NKT cell, or antibody-dependent cell-mediated cytotoxicity; ii) administration of NK1.1 specific IgG2a (mAb PK136) but not irrelevant IgG2a (myeloma M9144) prevented infection-induced B cell depletion consistent with a requirement for NK cells; iii) splenic NK cells but not T cells or NKT cells degranulated in infected C57BL/6 mice co-incident with B cell depletion evidenced by increased surface expression of CD107a; iv) purified NK cells from naïve C57BL/6 mice killed purified splenic B cells from T. brucei infected but not uninfected mice in vitro indicating acquisition of an NK cell activating phenotype by the post-infection B cells; v) adoptively transferred C57BL/6 NK cells prevented infection-induced B cell population growth in infected Prf1-/- mice consistent with in vivo B cell killing; vi) degranulated NK cells in infected mice had altered gene and differentiation antigen expression and lost cytotoxic activity consistent with functional exhaustion, but increased in number as infection progressed indicating continued generation. We conclude that NK cells in T. brucei infected mice kill B cells, suppress humoral immunity and expedite early mortality.
Collapse
Affiliation(s)
- Deborah Frenkel
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Fengqiu Zhang
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Patrick Guirnalda
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
| | - Carole Haynes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Viki Bockstal
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Stefan Magez
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- Laboratory for Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Structural Biology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Samuel J. Black
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
48
|
Lee H, Han NR, Hwang JY, Yun JI, Kim C, Park KH, Lee ST. Gelatin Directly Enhances Neurogenic Differentiation Potential in Bone Marrow-Derived Mesenchymal Stem Cells Without Stimulation of Neural Progenitor Cell Proliferation. DNA Cell Biol 2016; 35:530-6. [PMID: 27171118 DOI: 10.1089/dna.2016.3237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Gelatin has been reported to induce generation of mesenchymal stem cells (MSCs) with enhanced potential of differentiation into neuronal lineage cells. However, the presence of various cell types besides MSCs in bone marrow has raised doubts about the effects of gelatin. In the following report, we determined whether gelatin can directly enhance neurogenic differentiation potential in MSCs without proliferation of neural progenitor cells (NPCs). MSCs comprised a high proportion of bone marrow-derived primary cells (BMPCs) and gelatin induced significant increases in MSC proliferation during primary culture, and the proportion of MSCs was maintained at more than 99% throughout the subculture. However, NPCs comprised a low percentage of BMPCs and a decrease in proliferation was detected despite gelatin treatment during the primary culture, and the proportion of subcultured NPCs gradually decreased. In a similar manner, MSCs exposed to gelatin during primary culture showed more enhanced neurogenic differentiation ability than those not exposed to gelatin. Together, these results demonstrate that gelatin directly enhances neurogenic differentiation in bone marrow-derived MSCs without stimulating NPC proliferation.
Collapse
Affiliation(s)
- Hyun Lee
- 1 Department of Animal Life Science, Kangwon National University , Chuncheon, Korea
| | - Na Rae Han
- 1 Department of Animal Life Science, Kangwon National University , Chuncheon, Korea
| | - Jae Yeon Hwang
- 2 Division of Applied Animal Science, Kangwon National University , Chuncheon, Korea
| | - Jung Im Yun
- 3 Division of Animal Resource Science, Kangwon National University , Chuncheon, Korea
| | - Choonghyo Kim
- 4 Department of Neurosurgery, Kangwon National University Hospital, School of Medicine, Kangwon National University , Chuncheon, Korea
| | - Kyu Hyun Park
- 1 Department of Animal Life Science, Kangwon National University , Chuncheon, Korea.,3 Division of Animal Resource Science, Kangwon National University , Chuncheon, Korea
| | - Seung Tae Lee
- 1 Department of Animal Life Science, Kangwon National University , Chuncheon, Korea.,2 Division of Applied Animal Science, Kangwon National University , Chuncheon, Korea
| |
Collapse
|
49
|
Holodick NE, Vizconde T, Hopkins TJ, Rothstein TL. Age-Related Decline in Natural IgM Function: Diversification and Selection of the B-1a Cell Pool with Age. THE JOURNAL OF IMMUNOLOGY 2016; 196:4348-57. [PMID: 27183643 DOI: 10.4049/jimmunol.1600073] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/18/2016] [Indexed: 01/21/2023]
Abstract
Streptococcus pneumoniae is the most common cause of pneumonia, which claims the lives of people over the age of 65 y seven times more frequently than those aged 5-49 y. B-1a cells provide immediate and essential protection from S. pneumoniae through production of natural Ig, which has minimal insertion of N-region additions added by the enzyme TdT. In experiments with SCID mice infected with S. pneumoniae, we found passive transfer of IgG-depleted serum from aged (18-24 mo old) mice had no effect whereas IgG-depleted serum from young (3 mo old) mice was protective. This suggests protective natural IgM changes with age. Using single cell PCR we found N-region addition, which is initially low in fetal-derived B-1a cell IgM developing in the absence of TdT, increased in 7- to 24-mo-old mice as compared with 3-mo-old mice. To determine the mechanism responsible for the age related change in B-1a cell IgM, we established a mixed chimera system in which mice were reconstituted with allotype-marked mature peritoneal B-1a cells and adult bone marrow cells. We demonstrated even in the presence of mature peritoneal B-1a cells, adult bone marrow contributed to the mature B-1a cell pool. More importantly, using this system we found over a 10-mo-period peritoneal B-1a cell IgM changed, showing the number of cells lacking N-region additions at both junctions fell from 49 to 29% of sequences. These results strongly suggest selection-induced skewing alters B-1a cell-derived natural Ab, which may in turn be responsible for the loss of natural IgM-mediated protection against pneumococcal infection.
Collapse
Affiliation(s)
- Nichol E Holodick
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Manhasset, NY 11030;
| | - Teresa Vizconde
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Manhasset, NY 11030
| | - Thomas J Hopkins
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Manhasset, NY 11030
| | - Thomas L Rothstein
- Center for Oncology and Cell Biology, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Manhasset, NY 11030; Department of Medicine, Hofstra Northwell School of Medicine, Manhasset, NY 11030; and Department of Molecular Medicine, Hofstra Northwell School of Medicine, Manhasset, NY 11030
| |
Collapse
|
50
|
Popi AF, Longo-Maugéri IM, Mariano M. An Overview of B-1 Cells as Antigen-Presenting Cells. Front Immunol 2016; 7:138. [PMID: 27148259 PMCID: PMC4827000 DOI: 10.3389/fimmu.2016.00138] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/28/2016] [Indexed: 12/21/2022] Open
Abstract
The role of B cells as antigen-presenting cells (APCs) has been extensively studied, mainly in relation to the activation of memory T cells. Considering the B cell subtypes, the role of B-1 cells as APCs is beginning to be explored. Initially, it was described that B-1 cells are activated preferentially by T-independent antigens. However, some reports demonstrated that these cells are also involved in a T-dependent response. The aim of this review is to summarize information about the ability of B-1 cells to play a role as APCs and to briefly discuss the role of the BCR and toll-like receptor signals in this process. Furthermore, some characteristics of B-1 cells, such as natural IgM production and phagocytic ability, could interfere in the participation of these cells in the onset of an adaptive response.
Collapse
Affiliation(s)
- Ana F Popi
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo , São Paulo , Brazil
| | - Ieda M Longo-Maugéri
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo , São Paulo , Brazil
| | - Mario Mariano
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo , São Paulo , Brazil
| |
Collapse
|