1
|
Duarte ME, Deng Z, Kim SW. Effects of dietary Lactobacillus postbiotics and bacitracin on the modulation of mucosa-associated microbiota and pattern recognition receptors affecting immunocompetence of jejunal mucosa in pigs challenged with enterotoxigenic F18 + Escherichia coli. J Anim Sci Biotechnol 2024; 15:139. [PMID: 39390608 PMCID: PMC11468193 DOI: 10.1186/s40104-024-01098-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 09/01/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Enterotoxigenic Escherichia coli (E. coli) is a threat to humans and animals that causes intestinal disorders. Antimicrobial resistance has urged alternatives, including Lactobacillus postbiotics, to mitigate the effects of enterotoxigenic E. coli. METHODS Forty-eight newly weaned pigs were allotted to NC: no challenge/no supplement; PC: F18+ E. coli challenge/no supplement; ATB: F18+ E. coli challenge/bacitracin; and LPB: F18+ E. coli challenge/postbiotics and fed diets for 28 d. On d 7, pigs were orally inoculated with F18+ E. coli. At d 28, the mucosa-associated microbiota, immune and oxidative stress status, intestinal morphology, the gene expression of pattern recognition receptors (PRR), and intestinal barrier function were measured. Data were analyzed using the MIXED procedure in SAS 9.4. RESULTS PC increased (P < 0.05) Helicobacter mastomyrinus whereas reduced (P < 0.05) Prevotella copri and P. stercorea compared to NC. The LPB increased (P < 0.05) P. stercorea and Dialister succinatiphilus compared with PC. The ATB increased (P < 0.05) Propionibacterium acnes, Corynebacterium glutamicum, and Sphingomonas pseudosanguinis compared to PC. The PC tended to reduce (P = 0.054) PGLYRP4 and increased (P < 0.05) TLR4, CD14, MDA, and crypt cell proliferation compared with NC. The ATB reduced (P < 0.05) NOD1 compared with PC. The LPB increased (P < 0.05) PGLYRP4, and interferon-γ and reduced (P < 0.05) NOD1 compared with PC. The ATB and LPB reduced (P < 0.05) TNF-α and MDA compared with PC. CONCLUSIONS The F18+ E. coli challenge compromised intestinal health. Bacitracin increased beneficial bacteria showing a trend towards increasing the intestinal barrier function, possibly by reducing the expression of PRR genes. Lactobacillus postbiotics enhanced the immunocompetence of nursery pigs by increasing the expression of interferon-γ and PGLYRP4, and by reducing TLR4, NOD1, and CD14.
Collapse
Affiliation(s)
- Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, 116 Polk Hall, Campus Box 7621, Raleigh, NC, 27695, USA
| | - Zixiao Deng
- Department of Animal Science, North Carolina State University, 116 Polk Hall, Campus Box 7621, Raleigh, NC, 27695, USA
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, 116 Polk Hall, Campus Box 7621, Raleigh, NC, 27695, USA.
| |
Collapse
|
2
|
Ransegnola BP, Pattarabanjird T, McNamara CA. Tipping the Scale: Atheroprotective IgM-Producing B Cells in Atherosclerosis. Arterioscler Thromb Vasc Biol 2024; 44:1906-1915. [PMID: 39022832 PMCID: PMC11338718 DOI: 10.1161/atvbaha.124.319847] [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: 07/20/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease whose progression is fueled by proinflammatory moieties and limited by anti-inflammatory mediators. Whereas oxidative damage and the generation of oxidation-specific epitopes that act as damage-associated molecular patterns are highly inflammatory, IgM antibodies produced by B-1 and marginal zone B cells counteract unrestricted inflammation by neutralizing and encouraging clearance of these proinflammatory signals. In this review, we focus on describing the identities of IgM-producing B cells in both mice and humans, elaborating the mechanisms underlying IgM production, and discussing the potential strategies to augment the production of atheroprotective IgM. In addition, we will discuss promising therapeutic interventions in humans to help tip the scale toward augmentation of IgM production and to provide atheroprotection.
Collapse
Affiliation(s)
- Brett Patrick Ransegnola
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Department of Pathology, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tanyaporn Pattarabanjird
- Medical Scientist Training Program, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Coleen A. McNamara
- Beirne B. Carter Immunology Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Robert M. Berne Cardiovascular Research Center, Department of Medicine, University of Virginia, Charlottesville, VA, USA
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
3
|
Mattos MS, Vandendriessche S, Waisman A, Marques PE. The immunology of B-1 cells: from development to aging. Immun Ageing 2024; 21:54. [PMID: 39095816 PMCID: PMC11295433 DOI: 10.1186/s12979-024-00455-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
B-1 cells have intricate biology, with distinct function, phenotype and developmental origin from conventional B cells. They generate a B cell receptor with conserved germline characteristics and biased V(D)J recombination, allowing this innate-like lymphocyte to spontaneously produce self-reactive natural antibodies (NAbs) and become activated by immune stimuli in a T cell-independent manner. NAbs were suggested as "rheostats" for the chronic diseases in advanced age. In fact, age-dependent loss of function of NAbs has been associated with clinically-relevant diseases in the elderly, such as atherosclerosis and neurodegenerative disorders. Here, we analyzed comprehensively the ontogeny, phenotypic characteristics, functional properties and emerging roles of B-1 cells and NAbs in health and disease. Additionally, after navigating through the complexities of B-1 cell biology from development to aging, therapeutic opportunities in the field are discussed.
Collapse
Affiliation(s)
- Matheus Silvério Mattos
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Louvain, Belgium
| | - Sofie Vandendriessche
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Louvain, Belgium
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Centre of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Pedro Elias Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Louvain, Belgium.
| |
Collapse
|
4
|
Okabe Y. Development and organization of omental milky spots. Immunol Rev 2024; 324:68-77. [PMID: 38662554 DOI: 10.1111/imr.13337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/09/2024] [Indexed: 07/23/2024]
Abstract
The milky spots in omentum are atypical lymphoid tissues that play a pivotal role in regulating immune responses in the peritoneal cavity. The milky spots act as central hubs for collecting antigens and particles from the peritoneal cavity, regulating lymphocyte trafficking, promoting the differentiation and self-renewal of immune cells, and supporting the local germinal centre response. In addition, the milky spots exhibit unique developmental characteristics that combine the features of secondary and tertiary lymphoid tissues. These structures are innately programmed to form during foetal development; however, they can also be formed postnatally in response to peritoneal irritation such as inflammation, infection, obesity, or tumour metastasis. In this review, I discuss emerging perspectives on homeostatic development and organization of the milky spots.
Collapse
Affiliation(s)
- Yasutaka Okabe
- Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, Japan
| |
Collapse
|
5
|
Tan C, Reilly B, Ma G, Murao A, Jha A, Aziz M, Wang P. Neutrophils disrupt B-1a cell homeostasis by targeting Siglec-G to exacerbate sepsis. Cell Mol Immunol 2024; 21:707-722. [PMID: 38789529 PMCID: PMC11214631 DOI: 10.1038/s41423-024-01165-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/11/2024] [Indexed: 05/26/2024] Open
Abstract
B-1a cells, an innate-like cell population, are crucial for pathogen defense and the regulation of inflammation through their release of natural IgM and IL-10. In sepsis, B-1a cell numbers are decreased in the peritoneal cavity as they robustly migrate to the spleen. Within the spleen, migrating B-1a cells differentiate into plasma cells, leading to alterations in their original phenotype and functionality. We discovered a key player, sialic acid-binding immunoglobulin-like lectin-G (Siglec-G), which is expressed predominantly on B-1a cells and negatively regulates B-1a cell migration to maintain homeostasis. Siglec-G interacts with CXCR4/CXCL12 to modulate B-1a cell migration. Neutrophils aid B-1a cell migration via neutrophil elastase (NE)-mediated Siglec-G cleavage. Human studies revealed increased NE expression in septic patients. We identified an NE cleavage sequence in silico, leading to the discovery of a decoy peptide that protects Siglec-G, preserves peritoneal B-1a cells, reduces inflammation, and enhances sepsis survival. The role of Siglec-G in inhibiting B-1a cell migration to maintain their inherent phenotype and function is compromised by NE in sepsis, offering valuable insights into B-1a cell homeostasis. Employing a small decoy peptide to prevent NE-mediated Siglec-G cleavage has emerged as a promising strategy to sustain peritoneal B-1a cell homeostasis, alleviate inflammation, and ultimately improve outcomes in sepsis patients.
Collapse
Affiliation(s)
- Chuyi Tan
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Bridgette Reilly
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Gaifeng Ma
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Atsushi Murao
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Alok Jha
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA.
| | - Ping Wang
- Center for Immunology and Inflammation, the Feinstein Institutes for Medical Research, Manhasset, New York, USA.
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, USA.
| |
Collapse
|
6
|
Daley AD, Bénézech C. Fat-associated lymphoid clusters: Supporting visceral adipose tissue B cell function in immunity and metabolism. Immunol Rev 2024; 324:78-94. [PMID: 38717136 DOI: 10.1111/imr.13339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/09/2024] [Indexed: 07/23/2024]
Abstract
It is now widely understood that visceral adipose tissue (VAT) is a highly active and dynamic organ, with many functions beyond lipid accumulation and storage. In this review, we discuss the immunological role of this tissue, underpinned by the presence of fat-associated lymphoid clusters (FALCs). FALC's distinctive structure and stromal cell composition support a very different immune cell mix to that found in classical secondary lymphoid organs, which underlies their unique functions of filtration, surveillance, innate-like immune responses, and adaptive immunity within the serous cavities. FALCs are important B cell hubs providing B1 cell-mediated frontline protection against infection and supporting B2 cell-adaptative immune responses. Beyond these beneficial immune responses orchestrated by FALCs, immune cells within VAT play important homeostatic role. Dysregulation of immune cells during obesity and aging leads to chronic pathological "metabolic inflammation", which contributes to the development of cardiometabolic diseases. Here, we examine the emerging and complex functions of B cells in VAT homeostasis and the metabolic complications of obesity, highlighting the potential role that FALCs play and emphasize the areas where further research is needed.
Collapse
Affiliation(s)
- Alexander D Daley
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
7
|
Rodríguez-Zhurbenko N, Hernández AM. The role of B-1 cells in cancer progression and anti-tumor immunity. Front Immunol 2024; 15:1363176. [PMID: 38629061 PMCID: PMC11019000 DOI: 10.3389/fimmu.2024.1363176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/14/2024] [Indexed: 04/19/2024] Open
Abstract
In recent years, in addition to the well-established role of T cells in controlling or promoting tumor growth, a new wave of research has demonstrated the active involvement of B cells in tumor immunity. B-cell subsets with distinct phenotypes and functions play various roles in tumor progression. Plasma cells and activated B cells have been linked to improved clinical outcomes in several types of cancer, whereas regulatory B cells have been associated with disease progression. However, we are only beginning to understand the role of a particular innate subset of B cells, referred to as B-1 cells, in cancer. Here, we summarize the characteristics of B-1 cells and review their ability to infiltrate tumors. We also describe the potential mechanisms through which B-1 cells suppress anti-tumor immune responses and promote tumor progression. Additionally, we highlight recent studies on the protective anti-tumor function of B-1 cells in both mouse models and humans. Understanding the functions of B-1 cells in tumor immunity could pave the way for designing more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Nely Rodríguez-Zhurbenko
- Immunobiology Department, Immunology and Immunotherapy Division, Center of Molecular Immunology, Habana, Cuba
| | - Ana M. Hernández
- Applied Genetics Group, Department of Biochemistry, Faculty of Biology, University of Habana, Habana, Cuba
| |
Collapse
|
8
|
Ullah MA, Garcillán B, Whitlock E, Figgett WA, Infantino S, Eslami M, Yang S, Rahman MA, Sheng YH, Weber N, Schneider P, Tam CS, Mackay F. An unappreciated cell survival-independent role for BAFF initiating chronic lymphocytic leukemia. Front Immunol 2024; 15:1345515. [PMID: 38469292 PMCID: PMC10927009 DOI: 10.3389/fimmu.2024.1345515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/25/2024] [Indexed: 03/13/2024] Open
Abstract
Background Chronic Lymphocytic Leukemia (CLL) is characterized by the expansion of CD19+ CD5+ B cells but its origin remains debated. Mutated CLL may originate from post-germinal center B cells and unmutated CLL from CD5+ mature B cell precursors. Irrespective of precursor types, events initiating CLL remain unknown. The cytokines BAFF and APRIL each play a significant role in CLL cell survival and accumulation, but their involvement in disease initiation remains unclear. Methods We generated novel CLL models lacking BAFF or APRIL. In vivo experiments were conducted to explore the impact of BAFF or APRIL loss on leukemia initiation, progression, and dissemination. Additionally, RNA-seq and quantitative real-time PCR were performed to unveil the transcriptomic signature influenced by BAFF in CLL. The direct role of BAFF in controlling the expression of tumor-promoting genes was further assessed in patient-derived primary CLL cells ex-vivo. Results Our findings demonstrate a crucial role for BAFF, but not APRIL, in the initiation and dissemination of CLL cells. In the absence of BAFF or its receptor BAFF-R, the TCL1 transgene only increases CLL cell numbers in the peritoneal cavity, without dissemination into the periphery. While BAFF binding to BAFF-R is dispensable for peritoneal CLL cell survival, it is necessary to activate a tumor-promoting gene program, potentially linked to CLL initiation and progression. This direct role of BAFF in controlling the expression of tumor-promoting genes was confirmed in patient-derived primary CLL cells ex-vivo. Conclusions Our study, involving both mouse and human CLL cells, suggests that BAFF might initiate CLL through mechanisms independent of cell survival. Combining current CLL therapies with BAFF inhibition could offer a dual benefit by reducing peripheral tumor burden and suppressing transformed CLL cell output.
Collapse
Affiliation(s)
- Md Ashik Ullah
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - Beatriz Garcillán
- The Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Eden Whitlock
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - William A. Figgett
- The Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- Garvan Institute of Medical Research, Kinghorn Centre for Clinical Genomics, Darlinghurst, NSW, Australia
| | - Simona Infantino
- The Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Mahya Eslami
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Department of Oncology and Children’s Research Centre, University Children’s Hospital Zürich, Zürich, Switzerland
| | - SiLing Yang
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - M. Arifur Rahman
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - Yong H. Sheng
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - Nicholas Weber
- Cancer Care Services, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Constantine S. Tam
- Department of Haematology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Haematology, Monash University, Melbourne, VIC, Australia
| | - Fabienne Mackay
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
- The Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- The Department of Immunology and Pathology, Monash University, VIC, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
9
|
Lee EG, Oh JE. From neglect to spotlight: the underappreciated role of B cells in cutaneous inflammatory diseases. Front Immunol 2024; 15:1328785. [PMID: 38426103 PMCID: PMC10902158 DOI: 10.3389/fimmu.2024.1328785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
The skin, covering our entire body as its largest organ, manifests enormous complexities and a profound interplay of systemic and local responses. In this heterogeneous domain, B cells were considered strangers. Yet, recent studies have highlighted their existence in the skin and their distinct role in modulating cutaneous immunity across various immune contexts. Accumulating evidence is progressively shedding light on the significance of B cells in maintaining skin health and in skin disorders. Herein, we integrate current insights on the systemic and local contributions of B cells in three prevalent inflammatory skin conditions: Pemphigus Vulgaris (PV), Systemic Lupus Erythematosus (SLE), and Atopic Dermatitis (AD), underscoring the previously underappreciated importance of B cells within skin immunity. Moreover, we address the potential adverse effects of current treatments used for skin diseases, emphasizing their unintentional consequences on B cells. These comprehensive approaches may pave the way for innovative therapeutic strategies that effectively address the intricate nature of skin disorders.
Collapse
Affiliation(s)
- Eun-Gang Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji Eun Oh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
- BioMedical Research Center, KAIST, Daejeon, Republic of Korea
| |
Collapse
|
10
|
Maslanka J, Torres G, Londregan J, Goldman N, Silberman D, Somerville J, Riggs JE. Loss of B1 and marginal zone B cells during ovarian cancer. Cell Immunol 2024; 395-396:104788. [PMID: 38000306 PMCID: PMC10842900 DOI: 10.1016/j.cellimm.2023.104788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/31/2023] [Accepted: 11/20/2023] [Indexed: 11/26/2023]
Abstract
Recent advances in immunotherapy have not addressed the challenge presented by ovarian cancer. Although the peritoneum is an "accessible" locus for this disease there has been limited characterization of the immunobiology therein. We investigated the ID8-C57BL/6J ovarian cancer model and found marked depletion of B1 cells from the ascites of the peritoneal cavity. There was also selective loss of the B1 and marginal zone B cell subsets from the spleen. Immunity to antigens that activate these subsets validated their loss rather than relocation. A marked influx of myeloid-derived suppressor cells correlated with B cell subset depletion. These observations are discussed in the context of the housekeeping burden placed on innate B cells during ovarian cancer and to foster consideration of B cell biology in therapeutic strategies to address this challenge.
Collapse
Affiliation(s)
- Jeffrey Maslanka
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - Gretel Torres
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | | | - Naomi Goldman
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - Daniel Silberman
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - John Somerville
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA
| | - James E Riggs
- Department of Biology, Rider University, Lawrenceville, NJ 08648, USA.
| |
Collapse
|
11
|
Haas KM. Noncanonical B Cells: Characteristics of Uncharacteristic B Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1257-1265. [PMID: 37844278 PMCID: PMC10593487 DOI: 10.4049/jimmunol.2200944] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/12/2023] [Indexed: 10/18/2023]
Abstract
B lymphocytes were originally described as a cell type uniquely capable of secreting Abs. The importance of T cell help in Ab production was revealed soon afterward. Following these seminal findings, investigators made great strides in delineating steps in the conventional pathway that B cells follow to produce high-affinity Abs. These studies revealed generalized, or canonical, features of B cells that include their developmental origin and paths to maturation, activation, and differentiation into Ab-producing and memory cells. However, along the way, examples of nonconventional B cell populations with unique origins, age-dependent development, tissue localization, and effector functions have been revealed. In this brief review, features of B-1a, B-1b, marginal zone, regulatory, killer, NK-like, age-associated, and atypical B cells are discussed. Emerging work on these noncanonical B cells and functions, along with the study of their significance for human health and disease, represents an exciting frontier in B cell biology.
Collapse
Affiliation(s)
- Karen M Haas
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
12
|
Xie L, Wang H, Wu D, Zhang F, Chen W, Ye Y, Hu F. CXCL13 promotes thermogenesis in mice via recruitment of M2 macrophage and inhibition of inflammation in brown adipose tissue. Front Immunol 2023; 14:1253766. [PMID: 37936696 PMCID: PMC10627189 DOI: 10.3389/fimmu.2023.1253766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/10/2023] [Indexed: 11/09/2023] Open
Abstract
Introduction Brown adipose tissue (BAT) is mainly responsible for mammalian non-shivering thermogenesis and promotes energy expenditure. Meanwhile, similar to white adipose tissue (WAT), BAT also secretes a variety of adipokines to regulate metabolism through paracrine, autocrine, or endocrine ways. The chemokine C-X-C motif chemokine ligand-13 (CXCL13), a canonical B cell chemokine, functions in inflammation and tumor-related diseases. However, the role of CXCL13 in the adipose tissues is unclear. Methods The expression of CXCL13 in BAT and subcutaneous white adipose tissue (SWAT) of mice under cold stimulation were detected. Local injection of CXCL13 into BAT of normal-diet and high-fat-diet induced obese mice was used to detect thermogenesis and determine cold tolerance. The brown adipocytes were treated with CXCL13 alone or in the presence of macrophages to determine the effects of CXCL13 on thermogenic and inflammation related genes expression in vitro. Results In this study, we discovered that the expression of CXCL13 in the stromal cells of brown adipose tissue significantly elevated under cold stimulation. Overexpression of CXCL13 in the BAT via local injection could increase energy expenditure and promote thermogenesis in obese mice. Mechanically, CXCL13 could promote thermogenesis via recruiting M2 macrophages in the BAT and, in the meantime, inhibiting pro-inflammatory factor TNFα level. Discussion This study revealed the novel role of adipose chemokine CXCL13 in the regulation of BAT activity and thermogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fang Hu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, the Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
13
|
Castro PL, Barac F, Hansen TJ, Fjelldal PG, Hordvik I, Bjørgen H, Koppang EO. The Distribution of IgT mRNA + Cells in the Gut of the Atlantic Salmon ( Salmo salar L.). Animals (Basel) 2023; 13:3191. [PMID: 37893915 PMCID: PMC10603744 DOI: 10.3390/ani13203191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/01/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
The newly discovered IgT+ B cell is thought to play a dominant role in mucosal immunity, but limited studies have examined its distribution in fish species, hindering our understanding of its function. This study investigated IgT and poly Ig receptor (pIgR) mRNA+ cell distribution in Atlantic salmon (Salmo salar) gut using RNAscope in situ hybridization (ISH) and assessed the effects of vaccination. The pyloric caeca, mid-intestine (first and second parts), and posterior segment in two weight stages (Group 1: avg. 153 g, Group 2: avg. 1717 g) were examined in both vaccinated and unvaccinated fish. ISH revealed more IgT mRNA+ cells in the second part of the midgut compared to other intestinal segments, as well as a higher number of positive cells in Group 2 (older fish). In line with previous findings, intraperitoneal vaccination had no significant impact on the number of IgT+ transcripts. IgT mRNA+ cells were found mostly in the lamina propria and near capillaries, while pIgR was registered in both the lamina propria and mucosa. Interestingly, vaccinated fish presented adhesions and granulomatous tissue in the peritoneum, with both IgT and pIgR mRNA+ cells. Taken together, these results suggest that the distribution of IgT mRNA+ cells in the intestine of Atlantic salmon is region-specific and is not affected by intraperitoneal vaccination but varies with fish age.
Collapse
Affiliation(s)
- Pedro Luis Castro
- GIA-ECOAQUA, Universidad de Las Palmas de Gran Canaria, 35001 Telde, Spain
| | - Fran Barac
- Unit of Anatomy, Veterinary Faculty, Norwegian University of Life Sciences, 1433 Ås, Norway; (F.B.); (H.B.); (E.O.K.)
| | - Tom Johnny Hansen
- Matre Research Station, Institute of Marine Research, 5984 Matredal, Norway; (T.J.H.); (P.G.F.)
| | - Per Gunnar Fjelldal
- Matre Research Station, Institute of Marine Research, 5984 Matredal, Norway; (T.J.H.); (P.G.F.)
| | - Ivar Hordvik
- Institute of Biology, University of Bergen, 5007 Bergen, Norway;
| | - Håvard Bjørgen
- Unit of Anatomy, Veterinary Faculty, Norwegian University of Life Sciences, 1433 Ås, Norway; (F.B.); (H.B.); (E.O.K.)
| | - Erling Olaf Koppang
- Unit of Anatomy, Veterinary Faculty, Norwegian University of Life Sciences, 1433 Ås, Norway; (F.B.); (H.B.); (E.O.K.)
| |
Collapse
|
14
|
Suchanek O, Clatworthy MR. Homeostatic role of B-1 cells in tissue immunity. Front Immunol 2023; 14:1106294. [PMID: 37744333 PMCID: PMC10515722 DOI: 10.3389/fimmu.2023.1106294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/27/2023] [Indexed: 09/26/2023] Open
Abstract
To date, studies of tissue-resident immunity have mainly focused on innate immune cells and T cells, with limited data on B cells. B-1 B cells are a unique subset of B cells with innate-like properties, enriched in murine pleural and peritoneal cavities and distinct from conventional B-2 cells in their ontogeny, phenotype and function. Here we discuss how B-1 cells represent exemplar tissue-resident immune cells, summarizing the evidence for their long-term persistence & self-renewal within tissues, differential transcriptional programming shaped by organ-specific environmental cues, as well as their tissue-homeostatic functions. Finally, we review the emerging data supporting the presence and homeostatic role of B-1 cells across non-lymphoid organs (NLOs) both in mouse and human.
Collapse
Affiliation(s)
- Ondrej Suchanek
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Menna R. Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- NIHR Cambridge Biomedical Research Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
- Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
15
|
Ma K, Du W, Wang S, Xiao F, Li J, Tian J, Xing Y, Kong X, Rui K, Qin R, Zhu X, Wang J, Luo C, Wu H, Zhang Y, Wen C, He L, Liu D, Zou H, Lu Q, Wu L, Lu L. B1-cell-produced anti-phosphatidylserine antibodies contribute to lupus nephritis development via TLR-mediated Syk activation. Cell Mol Immunol 2023; 20:881-894. [PMID: 37291237 PMCID: PMC10250184 DOI: 10.1038/s41423-023-01049-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/22/2023] [Indexed: 06/10/2023] Open
Abstract
Autoantibodies produced by B cells play a pivotal role in the pathogenesis of systemic lupus erythematosus (SLE). However, both the cellular source of antiphospholipid antibodies and their contributions to the development of lupus nephritis (LN) remain largely unclear. Here, we report a pathogenic role of anti-phosphatidylserine (PS) autoantibodies in the development of LN. Elevated serum PS-specific IgG levels were measured in model mice and SLE patients, especially in those with LN. PS-specific IgG accumulation was found in the kidney biopsies of LN patients. Both transfer of SLE PS-specific IgG and PS immunization triggered lupus-like glomerular immune complex deposition in recipient mice. ELISPOT analysis identified B1a cells as the main cell type that secretes PS-specific IgG in both lupus model mice and patients. Adoptive transfer of PS-specific B1a cells accelerated the PS-specific autoimmune response and renal damage in recipient lupus model mice, whereas depletion of B1a cells attenuated lupus progression. In culture, PS-specific B1a cells were significantly expanded upon treatment with chromatin components, while blockade of TLR signal cascades by DNase I digestion and inhibitory ODN 2088 or R406 treatment profoundly abrogated chromatin-induced PS-specific IgG secretion by lupus B1a cells. Thus, our study has demonstrated that the anti-PS autoantibodies produced by B1 cells contribute to lupus nephritis development. Our findings that blockade of the TLR/Syk signaling cascade inhibits PS-specific B1-cell expansion provide new insights into lupus pathogenesis and may facilitate the development of novel therapeutic targets for the treatment of LN in SLE.
Collapse
Affiliation(s)
- Kongyang Ma
- Centre for Infection and Immunity Studies, School of Medicine, The Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, 999077, China
- Department of Rheumatology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Wenhan Du
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, 999077, China
- Chongqing International Institute for Immunology, Chongqing, 400038, China
| | - Shiyun Wang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, 999077, China
| | - Fan Xiao
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, 999077, China
| | - Jingyi Li
- Department of Rheumatology and Immunology, Southwest Hospital, The First Hospital Affiliated to Army Medical University, Chongqing, 400038, China
| | - Jie Tian
- Department of Laboratory Medicine, Affiliated Hospital and Institute of Medical Immunology, Jiangsu University, Zhenjiang, China
| | - Yida Xing
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaodan Kong
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ke Rui
- Department of Laboratory Medicine, Affiliated Hospital and Institute of Medical Immunology, Jiangsu University, Zhenjiang, China
| | - Rencai Qin
- Centre for Infection and Immunity Studies, School of Medicine, The Sun Yat-sen University, Shenzhen, 518107, Guangdong, China
| | - Xiaoxia Zhu
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Cainan Luo
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yun Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chengping Wen
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Lan He
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Dongzhou Liu
- Department of Rheumatology, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Hejian Zou
- Department of Rheumatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Lijun Wu
- Department of Rheumatology and Immunology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, 999077, China.
- Chongqing International Institute for Immunology, Chongqing, 400038, China.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong, China.
| |
Collapse
|
16
|
Gorskaya YF, Tukhvatulin AI, Dzharullaeva AS, Semenova EN, Vavilova LM, Nagurskaya EV, Bekhalo VA, Nesterenko VG. Effect of Activated Immunocompetent Cells on the Content of Multipotent Stromal Cells in Splenic Transplants of CBA and CBA/N Mice Administered with Polyvinylpyrrolidone. Bull Exp Biol Med 2023:10.1007/s10517-023-05813-2. [PMID: 37338771 DOI: 10.1007/s10517-023-05813-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Indexed: 06/21/2023]
Abstract
One day after intraperitoneal injection of polyvinylpyrrolidone (PVP) to recipient CBA and CBA/N mice, the count of multipotent stromal cells (MSC) in the 4-month-old splenic transplants was minimum in CBA/N→CBA/N group in comparison with the transplants of intact recipients (0.6 from the control level), but increased by 2.3, 3.2, and 3.7 times in CBA/N→CBA, CBA→CBA, and CBA→CBA/N groups, respectively. In the blood serum of recipient CBA/N mice with 4-month splenic transplants of CBA donors, the levels of some cytokines (IL-5, TNFα, and IL-2) was significantly increased 1 and 24 h after PVP injection in contrast to mice with bone marrow transplants, which attests to activation of the innate immunity mechanisms in this (splenic) transplantation variant. Probably, this phenomenon can be explained by the fact that the splenic transplants contain a sufficient number of CD+B-1a lymphocytes that can restore the response of recipient CBA/N mice to PVP. Thus, similar to bone marrow transplants [5], MSC count in splenic transplants increased only in groups, where the recipients were capable of responding to PVP. In other words, after injection of PVP to recipient mice, MSC counts in the spleen and bone marrow at this moment are determined by availability of activated immunocompetent cells. Overall, the novel data attest to close relationships between the stromal tissue of hematopoietic and lymphoid organs, on the one hand, and immune system, on the other.
Collapse
Affiliation(s)
- Yu F Gorskaya
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - A I Tukhvatulin
- Laboratory of Cellular Microbiology, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A Sh Dzharullaeva
- Laboratory of Cellular Microbiology, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E N Semenova
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - L M Vavilova
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - E V Nagurskaya
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V A Bekhalo
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V G Nesterenko
- Laboratory of Immunity Regulation and Immunologic Tolerance, N. F. Gamaleya National Research Center of Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
17
|
Figueiredo Galvao HB, Dinh QN, Thomas JM, Wassef F, Diep H, Bobik A, Sobey CG, Drummond GR, Vinh A. Proteasome inhibition reduces plasma cell and antibody secretion, but not angiotensin II-induced hypertension. Front Cardiovasc Med 2023; 10:1184982. [PMID: 37332591 PMCID: PMC10272792 DOI: 10.3389/fcvm.2023.1184982] [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: 03/13/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Depletion of mature B cells affords protection against experimental hypertension. However, whether B cell-mediated hypertension is dependent on differentiation into antibody-secreting cells (ASCs) remains unclear. Using the proteasome inhibitor, bortezomib, the present study tested the effect of ASC reduction on angiotensin II-induced hypertension. Methods Male C57BL6/J mice were infused with angiotensin II (0.7 mg/kg/day; s.c.) for 28 days via osmotic minipump to induce hypertension. Normotensive control mice received saline infusion. Bortezomib (750 μg/kg) or vehicle (0.1% DMSO) was administered (i.v.) 3 days prior to minipump implantation, and twice weekly thereafter. Systolic blood pressure was measured weekly using tail-cuff plethysmography. Spleen and bone marrow B1 (CD19+B220-), B2 (B220+CD19+) and ASCs (CD138hiSca-1+Blimp-1+) were enumerated by flow cytometry. Serum immunoglobulins were quantified using a bead-based immunoassay. Results Bortezomib treatment reduced splenic ASCs by ∼68% and ∼64% compared to vehicle treatment in normotensive (2.00 ± 0.30 vs. 0.64 ± 0.15 × 105 cells; n = 10-11) and hypertensive mice (0.52 ± 0.11 vs. 0.14 ± 0.02 × 105 cells; n = 9-11), respectively. Bone marrow ASCs were also reduced by bortezomib in both normotensive (4.75 ± 1.53 vs. 1.71 ± 0.41 × 103 cells; n = 9-11) and hypertensive mice (4.12 ± 0.82 vs. 0.89 ± 0.18 × 103 cells; n = 9-11). Consistent with ASC reductions, bortezomib reduced serum IgM and IgG2a in all mice. Despite these reductions in ASCs and antibody levels, bortezomib did not affect angiotensin II-induced hypertension over 28 days (vehicle: 182 ± 4 mmHg vs. bortezomib: 177 ± 7 mmHg; n = 9-11). Conclusion Reductions in ASCs and circulating IgG2a and IgM did not ameliorate experimental hypertension, suggesting other immunoglobulin isotypes or B cell effector functions may promote angiotensin II-induced hypertension.
Collapse
Affiliation(s)
- Hericka Bruna Figueiredo Galvao
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Quynh Nhu Dinh
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Jordyn M. Thomas
- Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Flavia Wassef
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Henry Diep
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Alex Bobik
- Baker Heart and Diabetes Institute, Prahran, Australia
- Department of Immunology, Monash University, Melbourne, VIC, Australia
- Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Christopher G. Sobey
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Prahran, Australia
| | - Grant R. Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Prahran, Australia
| | - Antony Vinh
- Department of Microbiology, Anatomy, Physiology and Pharmacology, Centre for Cardiovascular Biology and Disease Research, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
18
|
Spurrier MA, Jennings-Gee JE, Haas KM. Type I IFN Receptor Signaling on B Cells Promotes Antibody Responses to Polysaccharide Antigens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:148-157. [PMID: 36458995 PMCID: PMC9812919 DOI: 10.4049/jimmunol.2200538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/07/2022] [Indexed: 01/03/2023]
Abstract
We previously reported monophosphoryl lipid A (MPL) and synthetic cord factor trehalose-6,6'-dicorynomycolate (TDCM) significantly increase Ab responses to T cell-independent type 2 Ags (TI-2 Ags) in a manner dependent on B cell-intrinsic TLR4 expression, as well as MyD88 and TRIF proteins. Given the capacity of MPL to drive type I IFN production, we aimed to investigate the extent to which type I IFN receptor (IFNAR) signaling was required for TI-2 responses and adjuvant effects. Using Ifnar1-/- mice and IFNAR1 Ab blockade, we found that IFNAR signaling is required for optimal early B cell activation, expansion, and Ab responses to nonadjuvanted TI-2 Ags, including the pneumococcal vaccine. Further study demonstrated that B cell-intrinsic type I IFN signaling on B cells was essential for normal TI-2 Ab responses. In particular, TI-2 Ag-specific B-1b cell activation and expansion were significantly impaired in Ifnar1-/- mice; moreover, IFNAR1 Ab blockade similarly reduced activation, expansion, and differentiation of IFNAR1-sufficient B-1b cells in Ifnar1-/- recipient mice, indicating that B-1b cell-expressed IFNAR supports TI-2 Ab responses. Consistent with these findings, type I IFN significantly increased the survival of TI-2 Ag-activated B-1b cells ex vivo and promoted plasmablast differentiation. Nonetheless, MPL/TDCM adjuvant effects, which were largely carried out through innate B cells (B-1b and splenic CD23- B cells), were independent of type I IFN signaling. In summary, our study highlights an important role for B-1b cell-expressed IFNAR in promoting responses to nonadjuvanted TI-2 Ags, but it nonetheless demonstrates that adjuvants which support innate B cell responses may bypass this requirement.
Collapse
Affiliation(s)
- M Ariel Spurrier
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Jamie E Jennings-Gee
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC
| | - Karen M Haas
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
19
|
Gawish R, Maier B, Obermayer G, Watzenboeck ML, Gorki AD, Quattrone F, Farhat A, Lakovits K, Hladik A, Korosec A, Alimohammadi A, Mesteri I, Oberndorfer F, Oakley F, Brain J, Boon L, Lang I, Binder CJ, Knapp S. A neutrophil-B-cell axis impacts tissue damage control in a mouse model of intraabdominal bacterial infection via Cxcr4. eLife 2022; 11:e78291. [PMID: 36178806 PMCID: PMC9525059 DOI: 10.7554/elife.78291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is a life-threatening condition characterized by uncontrolled systemic inflammation and coagulation, leading to multiorgan failure. Therapeutic options to prevent sepsis-associated immunopathology remain scarce. Here, we established a mouse model of long-lasting disease tolerance during severe sepsis, manifested by diminished immunothrombosis and organ damage in spite of a high pathogen burden. We found that both neutrophils and B cells emerged as key regulators of tissue integrity. Enduring changes in the transcriptional profile of neutrophils include upregulated Cxcr4 expression in protected, tolerant hosts. Neutrophil Cxcr4 upregulation required the presence of B cells, suggesting that B cells promoted disease tolerance by improving tissue damage control via the suppression of neutrophils' tissue-damaging properties. Finally, therapeutic administration of a Cxcr4 agonist successfully promoted tissue damage control and prevented liver damage during sepsis. Our findings highlight the importance of a critical B-cell/neutrophil interaction during sepsis and establish neutrophil Cxcr4 activation as a potential means to promote disease tolerance during sepsis.
Collapse
Affiliation(s)
- Riem Gawish
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Barbara Maier
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Georg Obermayer
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of Laboratory Medicine, Medical University of ViennaViennaAustria
| | - Martin L Watzenboeck
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Anna-Dorothea Gorki
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Federica Quattrone
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Asma Farhat
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Karin Lakovits
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
| | - Anastasiya Hladik
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
| | - Ana Korosec
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
| | - Arman Alimohammadi
- Department of Medicine II, Division of Cardiology, Medical University of ViennaViennaAustria
| | - Ildiko Mesteri
- Department of Pathology, Medical University ViennaViennaAustria
| | | | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle UniversityNewcastleUnited Kingdom
| | - John Brain
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle UniversityNewcastleUnited Kingdom
| | | | - Irene Lang
- Department of Medicine II, Division of Cardiology, Medical University of ViennaViennaAustria
| | - Christoph J Binder
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of Laboratory Medicine, Medical University of ViennaViennaAustria
| | - Sylvia Knapp
- Department of Medicine I, Laboratory of Infection Biology, Medical University ViennaViennaAustria
- Ce-M-M-, Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| |
Collapse
|
20
|
Català C, Velasco-de Andrés M, Casadó-Llombart S, Leyton-Pereira A, Carrillo-Serradell L, Isamat M, Lozano F. Innate immune response to peritoneal bacterial infection. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 371:43-61. [PMID: 35965000 DOI: 10.1016/bs.ircmb.2022.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Spontaneous and secondary peritoneal infections, mostly of bacterial origin, easily spread to cause severe sepsis. Cellular and humoral elements of the innate immune system are constitutively present in peritoneal cavity and omentum, and play an important role in peritonitis progression and resolution. This review will focus on the description of the anatomic characteristics of the peritoneal cavity and the composition and function of such innate immune elements under both steady-state and bacterial infection conditions. Potential innate immune-based therapeutic interventions in bacterial peritonitis alternative or adjunctive to classical antibiotic therapy will be briefly discussed.
Collapse
Affiliation(s)
- Cristina Català
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Sergi Casadó-Llombart
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | - Marcos Isamat
- Sepsia Therapeutics S.L. 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francisco Lozano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Servei d'Immunologia, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Barcelona, Spain; Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
21
|
Chen H, Wang J, Zhang C, Ding P, Tian S, Chen J, Ji G, Wu T. Sphingosine 1-phosphate receptor, a new therapeutic direction in different diseases. Biomed Pharmacother 2022; 153:113341. [PMID: 35785704 DOI: 10.1016/j.biopha.2022.113341] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/01/2022] Open
Abstract
Sphingosine 1-phosphate receptor (S1PR), as a kind of G protein-coupled receptor, has five subtypes, including S1PR1, S1PR2, S1PR3, S1PR4, and S1PR5. Sphingosine 1-phosphate receptor (S1P) and S1PR regulate the trafficking of neutrophils and some cells, which has great effects on immune systems, lung tissue, and liver tissue. Presently, many related reports have proved that S1PR has a strong effect on the migration of lymphocytes, tumor cells, neutrophils, and many other cells via the regulation of signals, pathways, and enzymes. In this way, S1PR can regulate the relative response of the organism. Thus, S1PR has become a possible target for the treatment of autoimmune diseases, pulmonary disease, liver disease, and cancer. In this review, we mainly focus on the research of the S1PR for the new therapeutic directions of different diseases and is expected to assist support in the clinic and drug use.
Collapse
Affiliation(s)
- Hongyu Chen
- Minhang Hospital, Fudan University, Shanghai 201199, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Caiyun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peilun Ding
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuxia Tian
- Minhang Hospital, Fudan University, Shanghai 201199, China
| | - Junming Chen
- Minhang Hospital, Fudan University, Shanghai 201199, China.
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
22
|
Newell KL, Cox J, Waickman AT, Wilmore JR, Winslow GM. T-bet + B cells Dominate the Peritoneal Cavity B Cell Response during Murine Intracellular Bacterial Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2749-2760. [PMID: 35867676 PMCID: PMC9309898 DOI: 10.4049/jimmunol.2101209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/16/2022] [Indexed: 06/15/2023]
Abstract
T-bet+ B cells have emerged as a major B cell subset associated with both protective immunity and immunopathogenesis. T-bet is a transcription factor associated with the type I adaptive immune response to intracellular pathogens, driving an effector program characterized by the production of IFN-γ. Murine infection with the intracellular bacterium, Ehrlichia muris, generates protective extrafollicular T cell-independent T-bet+ IgM-secreting plasmablasts, as well as T-bet+ IgM memory cells. Although T-bet is a signature transcription factor for this subset, it is dispensable for splenic CD11c+ memory B cell development, but not for class switching to IgG2c. In addition to the T-bet+ plasmablasts found in the spleen, we show that Ab-secreting cells can also be found within the mouse peritoneal cavity; these cells, as well as their CD138- counterparts, also expressed T-bet. A large fraction of the T-bet+ peritoneal B cells detected during early infection were highly proliferative and expressed CXCR3 and CD11b, but, unlike in the spleen, they did not express CD11c. T-bet+ CD11b+ memory B cells were the dominant B cell population in the peritoneal cavity at 30 d postinfection, and although they expressed high levels of T-bet, they did not require B cell-intrinsic T-bet expression for their generation. Our data uncover a niche for T-bet+ B cells within the peritoneal cavity during intracellular bacterial infection, and they identify this site as a reservoir for T-bet+ B cell memory.
Collapse
Affiliation(s)
- Krista L Newell
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Justin Cox
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Adam T Waickman
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Joel R Wilmore
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| | - Gary M Winslow
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY
| |
Collapse
|
23
|
Abstract
Adipose tissue is a complex dynamic organ with whole-body immunometabolic influence. Much of the work into understanding the role of immune cells in adipose tissue has been in the context of obesity. These investigations have also uncovered a range of typical (immune) and non-typical functions exerted by adipose tissue leukocytes. Here we provide an overview of the adipose tissue immune system, including its role as an immune reservoir in the whole-body response to infection and as a site of parasitic and viral infections. We also describe the functional roles of specialized immunological structures found within adipose tissue. However, our main focus is on the recently discovered 'non-immune' functions of adipose tissue immune cells, which include the regulation of adipocyte homeostasis, as well as responses to changing nutrient status and body temperature. In doing so, we outline the therapeutic potential of the adipose tissue immune system in health and disease.
Collapse
|
24
|
Simón R, Martín-Martín A, Morel E, Díaz-Rosales P, Tafalla C. Functional and Phenotypic Characterization of B Cells in the Teleost Adipose Tissue. Front Immunol 2022; 13:868551. [PMID: 35619704 PMCID: PMC9127059 DOI: 10.3389/fimmu.2022.868551] [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] [Received: 02/02/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
The immune response of the adipose tissue (AT) has been neglected in most animal models until investigations in human and mice linked obesity to chronic inflammation, highlighting the immune nature of this tissue. Despite this, in teleost fish, only a few studies have addressed the immune role of the AT. These studies have mostly focused on reporting transcriptional changes in the AT in response to diverse intraperitoneally delivered stimuli. Although the presence of B cells within the AT was also previously revealed, these cells have never been phenotypically or functionally characterized and this is what we have addressed in the current study. Initially, the B cell populations present in the rainbow trout (Oncorhynchus mykiss) AT were characterized in comparison to B cells from other sources. As occurs in other rainbow trout tissues, IgM+IgD+, IgM+IgD- and IgD+IgM- B cell subsets were identified in the AT. Interestingly, AT IgM+IgD- B cells showed a transcriptional profile that agrees with that of cells that have committed to plasmablasts/plasma cells, being this profile much more pronounced towards a differentiation state than that of blood IgM+IgD- B cells. Accordingly, the IgM-secreting capacity of AT B cells is significantly higher than that of blood B cells. Additionally, AT IgM+IgD+ B cells also showed specific phenotypic traits when compared to their counterparts in other tissues. Finally, we established how these B cell subsets responded when rainbow trout were intraperitoneally injected with a model antigen. Our results demonstrate that the AT hosts plasmablasts/plasma cells that secrete specific IgMs, as happens in the peritoneal cavity and systemic immune tissues. Although the presence of these antigen-specific IgM-secreting cells was more abundant in the peritoneal cavity, these specific differentiated B cells were detected in the AT for long time periods at levels similar to those of spleen and head kidney. Our results provide new evidence regarding the immune role of the teleost AT, indicating that it functions as a secondary lymphoid organ that promotes immunity to peritoneal antigens.
Collapse
Affiliation(s)
- Rocío Simón
- Animal Health Research Center, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - Alba Martín-Martín
- Animal Health Research Center, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - Esther Morel
- Animal Health Research Center, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - Patricia Díaz-Rosales
- Animal Health Research Center, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| | - Carolina Tafalla
- Animal Health Research Center, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (CISA-INIA-CSIC), Madrid, Spain
| |
Collapse
|
25
|
She Z, Li C, Wu F, Mao J, Xie M, Hun M, Abdirahman AS, Luo S, Wan W, Tian J, Wen C. The Role of B1 Cells in Systemic Lupus Erythematosus. Front Immunol 2022; 13:814857. [PMID: 35418972 PMCID: PMC8995743 DOI: 10.3389/fimmu.2022.814857] [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] [Received: 11/14/2021] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by multisystemic and multi-organ involvement, recurrent relapses and remissions, and the presence of large amounts of autoantibodies in the body as the main clinical features. The mechanisms involved in this disease are complex and remain poorly understood; however, they are generally believed to be related to genetic susceptibility factors, external stimulation of the body’s immune dysfunction, and impaired immune regulation. The main immune disorders include the imbalance of T lymphocyte subsets, hyperfunction of B cells, production of large amounts of autoantibodies, and further deposition of immune complexes, which result in tissue damage. Among these, B cells play a major role as antibody-producing cells and have been studied extensively. B1 cells are a group of important innate-like immune cells, which participate in various innate and autoimmune processes. Yet the role of B1 cells in SLE remains unclear. In this review, we focus on the mechanism of B1 cells in SLE to provide new directions to explore the pathogenesis and treatment modalities of SLE.
Collapse
Affiliation(s)
- Zhou She
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cuifang Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Min Xie
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Marady Hun
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Amin Sheikh Abdirahman
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Senlin Luo
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wuqing Wan
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jidong Tian
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
26
|
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: 2] [Impact Index Per Article: 1.0] [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
|
27
|
Webster SE, Ryali B, Clemente MJ, Tsuji NL, Holodick NE. Sex Influences Age-Related Changes in Natural Antibodies and CD5 + B-1 Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1755-1771. [PMID: 35256511 PMCID: PMC8976758 DOI: 10.4049/jimmunol.2101150] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/30/2022] [Indexed: 11/19/2022]
Abstract
Natural Abs are primarily produced by B-1 cells and are essential for protection against Streptococcus pneumoniae The incidence and mortality rate for pneumococcal infection increases dramatically after age 65, disproportionately affecting males in both human and murine systems. To date, there is a significant gap in our understanding of the relationship among sex, aging, natural IgM efficacy, and the natural IgM repertoire. Our investigation demonstrates that the protective capacity of serum IgM against pneumococcal infection is maintained in IgM obtained from aged female mice but absent in IgM from aged male mice. To understand this difference in protective capacity, we examined serum Ig, discovering that the protective change was not associated with shifts in levels of phosphorylcholine (PC)- or pneumococcal capsular polysaccharide serotype 3-specific IgM. Interestingly, we observed that aged females have an increase in the total number of CD5+ B-1 cells, higher serum IL-5 levels, and a larger percentage of aged female CD5+ B-1 cells that express CD86 as compared with aged males. Furthermore, single-cell IgM repertoire analysis from peritoneal PC+, splenic PC+, and bone marrow CD5+ B-1 cell subsets demonstrated greater diversity with age and a higher level of germline status in female mice than previously observed in studies of aged male mice. Aged female CD5+ B-1 cells also expressed higher levels of transcripts associated with cell activity and self-renewal, such as Nanog and Hmga2 Taken together, these data indicate that females maintain a more diverse and active CD5+ B-1 cell pool and natural IgM repertoire, which has implications for sex-related susceptibility to infection and disease.
Collapse
Affiliation(s)
- Sarah E Webster
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI.,Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Brinda Ryali
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI.,Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI.,Department of Medicine, Rush University Medical Center, Chicago, IL; and
| | - Michael J Clemente
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI.,Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI.,Flow Cytometry and Imaging Core, Western Michigan Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Naomi L Tsuji
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI.,Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| | - Nichol E Holodick
- Center for Immunobiology, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI; .,Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI
| |
Collapse
|
28
|
Boontiam W, Hong J, Kitipongpysan S, Wattanachai S. Full-fat field cricket (Gryllus bimaculatus) as a substitute for fish meal and soybean meal for weaning piglets: effects on growth performance, intestinal health, and redox status. J Anim Sci 2022; 100:skac080. [PMID: 35289918 PMCID: PMC9047170 DOI: 10.1093/jas/skac080] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 03/11/2022] [Indexed: 11/30/2022] Open
Abstract
Full-fat field cricket meal (FCP) is an alternative protein ingredient in livestock production; however, the effects of replacing conventional protein sources with FCP in nursery diets have not been determined. In this study, the effects of the partial replacement of either fish meal or soybean meal with FCP on weaning pigs were evaluated, including the analyses of growth performance, nutrient utilization, intestinal morphology,
immunity, oxidative stress, and fecal microbial counts. A total of 100 crossbred weaning pigs [(Landrace × Large White) × Duroc] were allotted to one of the following five treatments with five replicates (four pigs/pen) and fed for 28 d postweaning. Treatments were 1) a corn-soybean meal (SBM)-based diet with 5% fish meal (Positive control; PC), 2) a corn-SBM-based diet without fish meal (Negative control; NC), 3) field crickets replacing fishmeal on a total Lys basis (FCP1), 4) field crickets replacing fishmeal on a kg/kg basis (FCP2), and 5) field crickets replacing fish meal and soybean meal (FCP3). The piglets on FCP1 had a higher body weight on days 14 and 28, and an increased average daily gain over the experimental period than NC (P < 0.05); FCP2 and FCP3 were similar to the FCP1 treatment. The incidence of diarrhea was lower under an FCP-supplemented diet than under the NC diet throughout the study (P < 0.05). Pigs fed FCP1 and FCP2 had a higher digestibility of crude
protein (P = 0.041), and all FCP groups increased crude fat digestibility (P = 0.024). FCP1 and FCP2 also increased jejunal villus height
(P = 0.009), whereas the increase in jejunal villus-to-crypt ratios (P = 0.019) was greater in pigs fed the FCP2 diet than those fed the NC diet. Furthermore, FCP2 supplementation increased serum immunoglobulin A levels on days 14 and 28, including reduced serum interleukin-6 and tumor necrosis factor alpha levels (P < 0.05). Pigs fed an FCP2 diet had reduced malondialdehyde levels than those fed a PC diet, while pigs fed an FCP2 diet had higher superoxide dismutase and glutathione peroxidase levels, and more fecal Lactobacillus spp. than those fed an NC diet (P < 0.05). These results support the use of FCP as an alternative protein ingredient with beneficial effects on growth performance, intestinal morphology, antioxidant capacity, and intestinal microbiota. In particular, FCP can be used as a partial substitute for fish meal and soybean meal without detrimental effects on weaning pigs.
Collapse
Affiliation(s)
- Waewaree Boontiam
- Division of Animal Science, Faculty of Agriculture, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Jinsu Hong
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, U083SA, USA
| | - Sumetee Kitipongpysan
- Division of Agriculture, Faculty of Agriculture and Life Science, Chandrakasem Rajabhat University, Bangkok 10900, Thailand
| | - Suchat Wattanachai
- Division of Surgery and Theriogenology, Faculty of Veterinary Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
29
|
Halperin ST, ’t Hart BA, Luchicchi A, Schenk GJ. The Forgotten Brother: The Innate-like B1 Cell in Multiple Sclerosis. Biomedicines 2022; 10:606. [PMID: 35327408 PMCID: PMC8945227 DOI: 10.3390/biomedicines10030606] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system (CNS), traditionally considered a chronic autoimmune attack against the insulating myelin sheaths around axons. However, the exact etiology has not been identified and is likely multi-factorial. Recently, evidence has been accumulating that implies that autoimmune processes underlying MS may, in fact, be triggered by pathological processes initiated within the CNS. This review focuses on a relatively unexplored immune cell-the "innate-like" B1 lymphocyte. The B1 cell is a primary-natural-antibody- and anti-inflammatory-cytokine-producing cell present in the healthy brain. It has been recently shown that its frequency and function may differ between MS patients and healthy controls, but its exact involvement in the MS pathogenic process remains obscure. In this review, we propose that this enigmatic cell may play a more prominent role in MS pathology than ever imagined. We aim to shed light on the human B1 cell in health and disease, and how dysregulation in its delicate homeostatic role could impact MS. Furthermore, novel therapeutic avenues to restore B1 cells' beneficial functions will be proposed.
Collapse
Affiliation(s)
| | | | - Antonio Luchicchi
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HZ Amsterdam, The Netherlands; (S.T.H.); (B.A.’t.H.)
| | - Geert J. Schenk
- Department of Anatomy and Neurosciences, Amsterdam Neuroscience, MS Center Amsterdam, Amsterdam UMC, Vrije Universiteit, 1081 HZ Amsterdam, The Netherlands; (S.T.H.); (B.A.’t.H.)
| |
Collapse
|
30
|
Barington L, Christensen LVV, Pedersen KK, Niss Arfelt K, Roumain M, Jensen KHR, Kjær VMS, Daugvilaite V, Kearney JF, Christensen JP, Hjortø GM, Muccioli GG, Holst PJ, Rosenkilde MM. GPR183 Is Dispensable for B1 Cell Accumulation and Function, but Affects B2 Cell Abundance, in the Omentum and Peritoneal Cavity. Cells 2022; 11:cells11030494. [PMID: 35159303 PMCID: PMC8834096 DOI: 10.3390/cells11030494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
B1 cells constitute a specialized subset of B cells, best characterized in mice, which is abundant in body cavities, including the peritoneal cavity. Through natural and antigen-induced antibody production, B1 cells participate in the early defense against bacteria. The G protein-coupled receptor 183 (GPR183), also known as Epstein-Barr virus-induced gene 2 (EBI2), is an oxysterol-activated chemotactic receptor that regulates migration of B cells. We investigated the role of GPR183 in B1 cells in the peritoneal cavity and omentum. B1 cells expressed GPR183 at the mRNA level and migrated towards the GPR183 ligand 7α,25-dihydroxycholesterol (7α,25-OHC). GPR183 knock-out (KO) mice had smaller omenta, but with normal numbers of B1 cells, whereas they had fewer B2 cells in the omentum and peritoneal cavity than wildtype (WT) mice. GPR183 was not responsible for B1 cell accumulation in the omentum in response to i.p. lipopolysaccharide (LPS)-injection, in spite of a massive increase in 7α,25-OHC levels. Lack of GPR183 also did not affect B1a- or B1b cell-specific antibody responses after vaccination. In conclusion, we found that GPR183 is non-essential for the accumulation and function of B1 cells in the omentum and peritoneal cavity, but that it influences the abundance of B2 cells in these compartments.
Collapse
Affiliation(s)
- Line Barington
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
| | - Liv von Voss Christensen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
| | - Kristian Kåber Pedersen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
| | - Kristine Niss Arfelt
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
| | - Martin Roumain
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Brussels, Belgium; (M.R.); (G.G.M.)
| | - Kristian Høj Reveles Jensen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
| | - Viktoria Madeline Skovgaard Kjær
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
| | - Viktorija Daugvilaite
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
| | - John F. Kearney
- Division of Developmental and Clinical Immunology, Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Jan Pravsgaard Christensen
- Infectious Immunology Group, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Gertrud Malene Hjortø
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Brussels, Belgium; (M.R.); (G.G.M.)
| | - Peter Johannes Holst
- Experimental Vaccinology Group, Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
- InProTher ApS, 2200 Copenhagen, Denmark
| | - Mette Marie Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.B.); (L.v.V.C.); (K.K.P.); (K.N.A.); (K.H.R.J.); (V.M.S.K.); (V.D.); (G.M.H.)
- Correspondence:
| |
Collapse
|
31
|
Duarte ME, Kim SW. Intestinal microbiota and its interaction to intestinal health in nursery pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2022; 8:169-184. [PMID: 34977387 PMCID: PMC8683651 DOI: 10.1016/j.aninu.2021.05.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023]
Abstract
The intestinal microbiota has gained increased attention from researchers within the swine industry due to its role in promoting intestinal maturation, immune system modulation, and consequently the enhancement of the health and growth performance of the host. This review aimed to provide updated scientific information on the interaction among intestinal microbiota, dietary components, and intestinal health of pigs. The small intestine is a key site to evaluate the interaction of the microbiota, diet, and host because it is the main site for digestion and absorption of nutrients and plays an important role within the immune system. The diet and its associated components such as feed additives are the main factors affecting the microbial composition and is central in stimulating a beneficial population of microbiota. The microbiota–host interaction modulates the immune system, and, concurrently, the immune system helps to modulate the microbiota composition. The direct interaction between the microbiota and the host is an indication that the mucosa-associated microbiota can be more effective in evaluating its effect on health parameters. It was demonstrated that the mucosa-associated microbiota should be evaluated when analyzing the interaction among diets, microbiota, and health. In addition, supplementation of feed additives aimed to promote the intestinal health of pigs should consider their roles in the modulation of mucosa-associated microbiota as biomarkers to predict the response of growth performance to dietary interventions.
Collapse
Affiliation(s)
- Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, United States
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC, 27695, United States
| |
Collapse
|
32
|
Mouse innate-like B-1 lymphocytes promote inhaled particle-induced in vitro granuloma formation and inflammation in conjunction with macrophages. Arch Toxicol 2021; 96:585-599. [PMID: 34935064 PMCID: PMC8837577 DOI: 10.1007/s00204-021-03200-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/09/2021] [Indexed: 11/20/2022]
Abstract
The current paradigm for explaining lung granulomatous diseases induced by inhaled particles is mainly based on macrophages. This mechanism is now challenging because B lymphocytes also infiltrate injured tissue, and the deficiency in B lymphocytes is associated with limited lung granulomas in silica-treated mice. Here, we investigated how B lymphocytes respond to micro- and nanoparticles by combining in vivo and in vitro mouse models. We first demonstrated that innate-like B-1 lymphocytes (not conventional B-2 lymphocytes or plasma cells) specifically accumulated during granuloma formation in mice instilled with crystalline silica (DQ12, 2.5 mg/mouse) and carbon nanotubes (CNT Mitsui, 0.2 mg/mouse). In comparison to macrophages, peritoneal B-1 lymphocytes purified from naïve mice were resistant to the pyroptotic activity of reactive particles (up to 1 mg/mL) but clustered to establish in vitro cell/particle aggregates. Mouse B-1 lymphocytes (not B-2 lymphocytes) in coculture with macrophages and CNT (0.1 µg/mL) organized three-dimensional spheroid structures in Matrigel and stimulated the release of TIMP-1. Furthermore, purified B-1 lymphocytes are sensitive to nanosilica toxicity through radical generation in culture. Nanosilica-exposed B-1 lymphocytes released proinflammatory cytokines and alarmins. In conclusion, our data indicate that in addition to macrophages, B-1 lymphocytes participate in micrometric particle-induced granuloma formation and display inflammatory functions in response to nanoparticles.
Collapse
|
33
|
Souza SP, Splitt SD, Sànchez-Arcila JC, Alvarez JA, Wilson JN, Wizzard S, Luo Z, Baumgarth N, Jensen KDC. Genetic mapping reveals Nfkbid as a central regulator of humoral immunity to Toxoplasma gondii. PLoS Pathog 2021; 17:e1010081. [PMID: 34871323 PMCID: PMC8675933 DOI: 10.1371/journal.ppat.1010081] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 12/16/2021] [Accepted: 11/01/2021] [Indexed: 12/29/2022] Open
Abstract
Protective immunity to parasitic infections has been difficult to elicit by vaccines. Among parasites that evade vaccine-induced immunity is Toxoplasma gondii, which causes lethal secondary infections in chronically infected mice. Here we report that unlike susceptible C57BL/6J mice, A/J mice were highly resistant to secondary infection. To identify correlates of immunity, we utilized forward genetics to identify Nfkbid, a nuclear regulator of NF-κB that is required for B cell activation and B-1 cell development. Nfkbid-null mice (“bumble”) did not generate parasite-specific IgM and lacked robust parasite-specific IgG, which correlated with defects in B-2 cell maturation and class-switch recombination. Though high-affinity antibodies were B-2 derived, transfer of B-1 cells partially rescued the immunity defects observed in bumble mice and were required for 100% vaccine efficacy in bone marrow chimeric mice. Immunity in resistant mice correlated with robust isotype class-switching in both B cell lineages, which can be fine-tuned by Nfkbid gene expression. We propose a model whereby humoral immunity to T. gondii is regulated by Nfkbid and requires B-1 and B-2 cells for full protection. Eukaryotic parasitic diseases account for approximately one fifth of all childhood deaths, yet no highly protective vaccine exists for any human parasite. More research must be done to discover how to elicit protective vaccine-induced immunity to parasitic pathogens. We used an unbiased genetic screen to find key genes responsible for immunity to the eukaryotic parasite Toxoplasma gondii. Our screen found Nfkbid, a transcription factor regulator, which controls B cell activation and innate-like B-1 cell development. Mice without Nfkbid were not protected against T. gondii and were deficient at making antibodies against the parasite. Our survival studies of vaccinated mice with and without B-1 compartments found that B-1 cells improved survival, suggesting that B-1 cells act in conjunction with B-2 cells to provide vaccine-induced immunity. Nfkbid and other loci identified in our unbiased screen represent potential targets for vaccines to elicit protective immune responses against parasitic pathogens.
Collapse
Affiliation(s)
- Scott P. Souza
- School of Natural Sciences, Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
- Graduate Program in Quantitative and Systems Biology, University of California, Merced, Merced, California, United States of America
| | - Samantha D. Splitt
- School of Natural Sciences, Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
- Graduate Program in Quantitative and Systems Biology, University of California, Merced, Merced, California, United States of America
| | - Juan C. Sànchez-Arcila
- School of Natural Sciences, Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
| | - Julia A. Alvarez
- School of Natural Sciences, Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
- Graduate Program in Quantitative and Systems Biology, University of California, Merced, Merced, California, United States of America
| | - Jessica N. Wilson
- School of Natural Sciences, Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
- Graduate Program in Quantitative and Systems Biology, University of California, Merced, Merced, California, United States of America
| | - Safuwra Wizzard
- School of Natural Sciences, Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
| | - Zheng Luo
- Center for Immunology & Infectious Diseases, and Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, California, United States of America
| | - Nicole Baumgarth
- Center for Immunology & Infectious Diseases, and Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, California, United States of America
| | - Kirk D. C. Jensen
- School of Natural Sciences, Department of Molecular and Cell Biology, University of California, Merced, Merced, California, United States of America
- Health Science Research Institute, University of California, Merced, Merced, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Abstract
There are numbers of leukocytes present in peritoneal cavity, not only protecting body cavity from infection but also contributing to peripheral immunity including natural antibody production in circulation. The peritoneal leukocytes compose unique immune compartment, the functions of which cannot be replaced by other lymphoid organs. Atypical lymphoid clusters, called "milky spots", that are located in visceral adipose tissue omentum have the privilege of immune niche in terms of differentiation, recruitment, and activation of peritoneal immunity, yet mechanisms underlying the regulation are underexplored. In this review, I discuss the emerging views of peritoneal immune system in the contexts of its development, organization, and functions.
Collapse
Affiliation(s)
- Yasutaka Okabe
- Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center Osaka University, 3-1 Yamada-oka, Suita, 565-0871, Osaka, Japan.
| |
Collapse
|
35
|
Cheng YC, Duarte ME, Kim SW. Nutritional and functional values of lysed Corynebacterium glutamicum cell mass for intestinal health and growth of nursery pigs. J Anim Sci 2021; 99:skab331. [PMID: 34902029 PMCID: PMC8668180 DOI: 10.1093/jas/skab331] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/15/2021] [Indexed: 12/16/2022] Open
Abstract
The objective was to determine the nutritional and functional values of lysed Corynebacterium glutamicum cell mass (CGCM) as a protein supplement and a source of cell wall fragments supporting the growth and intestinal health of nursery pigs. Thirty-two pigs (21 d of age) were allotted to four treatments (n = 8) based on the randomized block design with sex and initial body weight (BW) as blocks. The main effect was the dietary supplementation of lysed CGCM (0, 0.7, 1.4, and 2.1%) replacing blood plasma and fed in two phases (10 and 11 d, respectively). Feed intake and BW were measured at the end of each phase. Pigs were euthanized on day 21 to collect jejunal tissue and mucosa to evaluate intestinal health. Ileal digesta were collected to measure the apparent ileal digestibility of nutrients in diets. Data were analyzed using Proc Mixed and Reg of SAS. Increasing daily intake of CGCM increased (linear; P < 0.05) ADG of pigs. Increasing CGCM supplementation affected (quadratic; P < 0.05) the relative abundance of Lactobacillaceae (minimum: 26.4% at 1.2% CGCM), Helicobacteraceae (maximum: 29.3% at 1.2% CGCM), and Campylobacteraceae (maximum: 9.0% at 1.0% CGCM). Increasing CGCM supplementation affected (quadratic; P < 0.05) the concentrations of immunoglobulin G (maximum: 4.94 µg/mg of protein at 1.0% CGCM) and protein carbonyl (PC; maximum: 6.12 nmol/mg of protein at 1.1% CGCM), whereas linearly decreased (P < 0.05) malondialdehyde (MDA) in the proximal jejunal mucosa. Increasing CGCM supplemention affected (quadratic; P < 0.05) intestinal enterocyte proliferation rate (maximum: 13.3% at 1.0% CGCM), whereas it did not affect intestinal morphology and the nutrient digestibility. In conclusion, supplementing 1.0% to 1.2%, reducing blood plasma supplementation by 0.7% to 0.9%, respectively, increased potential pathogenic microbiota associated in the jejunal mucosa resulting in increased immune response, enterocyte proliferation, and PC concentration. However, supplementing diets with 2.1% CGCM, replacing 1.5% blood plasma, improved growth performance, and reduced MDA without affecting nutrient digestibility, intestinal morphology, and microbiota in the jejunal mucosa. In this study, based on the polynomial contrast, supplementing 1.0% to 1.2% CGCM suppressed the benefits from blood plasma, whereas supplementing 2.1% CGCM showed functional benefits of CGCM with similar effects from blood plasma supplementation.
Collapse
Affiliation(s)
- Yi-Chi Cheng
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Marcos Elias Duarte
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
36
|
Mycobiota-induced IgA antibodies regulate fungal commensalism in the gut and are dysregulated in Crohn's disease. Nat Microbiol 2021; 6:1493-1504. [PMID: 34811531 PMCID: PMC8622360 DOI: 10.1038/s41564-021-00983-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022]
Abstract
Secretory immunoglobulin A (sIgA) plays an important role in gut barrier protection by shaping the resident microbiota community, restricting the growth of bacterial pathogens and enhancing host protective immunity via immunological exclusion. Here, we found that a portion of the microbiota-driven sIgA response is induced by and directed towards intestinal fungi. Analysis of the human gut mycobiota bound by sIgA revealed a preference for hyphae, a fungal morphotype associated with virulence. Candida albicans was a potent inducer of IgA class-switch recombination among plasma cells, via an interaction dependent on intestinal phagocytes and hyphal programming. Characterization of sIgA affinity and polyreactivity showed that hyphae-associated virulence factors were bound by these antibodies and that sIgA influenced C. albicans morphotypes in the murine gut. Furthermore, an increase in granular hyphal morphologies in patients with Crohn's disease compared with healthy controls correlated with a decrease in antifungal sIgA antibody titre with affinity to two hyphae-associated virulence factors. Thus, in addition to its importance in gut bacterial regulation, sIgA targets the uniquely fungal phenomenon of hyphal formation. Our findings indicate that antifungal sIgA produced in the gut can play a role in regulating intestinal fungal commensalism by coating fungal morphotypes linked to virulence, thereby providing a protective mechanism that might be dysregulated in patients with Crohn's disease.
Collapse
|
37
|
Chuah JJM, Hertzog PJ, Campbell NK. Immunoregulation by type I interferons in the peritoneal cavity. J Leukoc Biol 2021; 111:337-353. [PMID: 34612523 DOI: 10.1002/jlb.3mr0821-147r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The peritoneal cavity, a fluid-containing potential space surrounding the abdominal and pelvic organs, is home to a rich network of immune cells that maintain tissue homeostasis and provide protection against infection. However, under pathological conditions such as peritonitis, endometriosis, and peritoneal carcinomatosis, the peritoneal immune system can become dysregulated, resulting in nonresolving inflammation and disease progression. An enhanced understanding of the factors that regulate peritoneal immune cells under both homeostatic conditions and in disease contexts is therefore required to identify new treatment strategies for these often life-limiting peritoneal pathologies. Type I interferons (T1IFNs) are a family of cytokines with broad immunoregulatory functions, which provide defense against viruses, bacteria, and cancer. There have been numerous reports of immunoregulation by T1IFNs within the peritoneal cavity, which can contribute to both the resolution or propagation of peritoneal disease states, depending on the specifics of the disease setting and local environment. In this review, we provide an overview of the major immune cell populations that reside in the peritoneal cavity (or infiltrate it under inflammatory conditions) and highlight their contribution to the initiation, progression, or resolution of peritoneal diseases. Additionally, we will discuss the role of T1IFNs in the regulation of peritoneal immune cells, and summarize the results of laboratory studies and clinical trials which have investigated T1IFNs in peritonitis/sepsis, endometriosis, and peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Jasmine J M Chuah
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Nicole K Campbell
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
38
|
Barbosa CHD, Lantier L, Reynolds J, Wang J, Re F. Critical role of IL-25-ILC2-IL-5 axis in the production of anti-Francisella LPS IgM by B1 B cells. PLoS Pathog 2021; 17:e1009905. [PMID: 34449811 PMCID: PMC8428711 DOI: 10.1371/journal.ppat.1009905] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/09/2021] [Accepted: 08/17/2021] [Indexed: 01/21/2023] Open
Abstract
B1 cells, a subset of B lymphocytes whose developmental origin, phenotype, and function differ from that of conventional B2 cells, are the main source of “natural” IgM but can also respond to infection by rapidly producing pathogen-specific IgM directed against T-independent antigens. Francisella tularensis (Ft) is a Gram-negative bacterium that causes tularemia. Infection with Ft Live Vaccine Strain activates B1 cells for production of IgM directed against the bacterial LPS in a process incompletely understood. Here we show that immunization with purified Ft LPS elicits production of LPS-specific IgM and IgG3 by B1 cells independently of TLR2 or MyD88. Immunization, but not infection, generated peritoneum-resident memory B1 cells that differentiated into LPS-specific antibody secreting cells (ASC) upon secondary challenge. IL-5 was rapidly induced by immunization with Ft LPS and was required for production of LPS-specific IgM. Antibody-mediated depletion of ILC2 indicated that these cells were the source of IL-5 and were required for IgM production. IL-25, an alarmin that strongly activates ILC2, was rapidly secreted in response to immunization or infection and its administration to mice significantly increased IgM production and B1 cell differentiation to ASC. Conversely, mice lacking IL-17RB, the IL-25 receptor, showed impaired IL-5 induction, IgM production, and B1 ASC differentiation in response to immunization. Administration of IL-5 to Il17rb-/- mice rescued these B1 cells-mediated responses. Il17rb-/- mice were more susceptible to infection with Ft LVS and failed to develop immunity upon secondary challenge suggesting that LPS-specific IgM is one of the protective adaptive immune mechanisms against tularemia. Our results indicated that immunization with Ft LPS triggers production of IL-25 that, through stimulation of IL-5 release by ILC2, promotes B1 cells activation and differentiation into IgM secreting cells. By revealing the existence of an IL-25-ILC2-IL-5 axis our results suggest novel strategies to improve vaccination against T-independent bacterial antigens. B1 cells are a subset of B lymphocytes that participate in the immune response to infection by producing antibodies of the IgM class. Here we investigate the mechanisms that control B1 cells activation and production of IgM directed against the lipopolysaccharide (LPS) of Francisella tularensis, a Gram-negative bacterium that causes tularemia. Using a mouse model of tularemia, our results revealed that Francisella LPS elicits production of the cytokine IL-25 that in turn activates blood cells called Innate Lymphoid Cells 2 (ILC2). Once activated, ILC2 produce the cytokine IL-5 that is required for activation of B1 cells and production of IgM. Mice unresponsive to IL-25 are more susceptible to F. tularensis infection. By revealing the existence of an IL-25-ILC2-IL-5 axis our results suggest novel strategies to improve vaccination against bacteria.
Collapse
Affiliation(s)
- Carlos Henrique D. Barbosa
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Louis Lantier
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Joseph Reynolds
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Jinyong Wang
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Fabio Re
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
39
|
Royster W, Ochani M, Aziz M, Wang P. Therapeutic Potential of B-1a Cells in Intestinal Ischemia-reperfusion Injury. J Surg Res 2021; 268:326-336. [PMID: 34399355 DOI: 10.1016/j.jss.2021.06.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/14/2021] [Accepted: 06/29/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Acute mesenteric ischemia is a common surgical emergency. Restoration of blood flow is a critical objective of treating this pathology. However, many patients suffer from ischemia-reperfusion (I/R) injuries at the time of revascularization, requiring prolonged hospitalizations. B-1a cells are a subtype of B lymphocytes with roles in regulating inflammation and tissue injury by spontaneous release of natural IgM and IL-10. We hypothesized that treatment with B-1a cells protects mice from intestinal I/R. METHODS Mesenteric ischemia was induced in mice by placing a vascular clip on the superior mesenteric artery for 60 minutes. At the time of reperfusion, B-1a cells or PBS control were instilled into the peritoneal cavity (PerC) of mice. PerC lavage, blood, intestine, and lungs were collected 4 h after reperfusion. Serum organ injury and inflammatory markers such as ALT, AST, LDH, lactate, IL-6, as well as lung and gut histology and myeloperoxidase (MPO) were assessed. RESULTS In intestinal I/R, B-1a cell frequency and number in the PerC were significantly decreased compared to sham-operated mice. There was an increase in the serum levels of ALT, AST, LDH, lactate, and IL-6 when comparing the vehicle group with the sham group. These increases were significantly reduced in the B-1a cell treated group. B-1a cell treatment significantly decreased the intestine and lung injury scores as well as MPO content, compared to vehicle treated mice. B-1a cell treatment resulted in a reduction of apoptotic cells in these tissues. Serum IgM levels were decreased in intestinal I/R, while treatment with B-1a cells significantly increased their levels towards normal levels. CONCLUSIONS B-1a cell treatment at the time of mesenteric reperfusion ameliorates end organ damage and reduces systemic inflammation through the improvement of serum IgM levels. Preserving B-1a cells pool could serve as a novel therapeutic avenue in intestinal I/R injury.
Collapse
Affiliation(s)
- William Royster
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York; Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, Manhasset, New York
| | - Mahendar Ochani
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York; Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, Manhasset, New York
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York; Elmezzi Graduate School of Molecular Medicine, Manhasset, New York; Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, Manhasset, New York; Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York.
| |
Collapse
|
40
|
Rempenault C, Mielle J, Schreiber K, Corbeau P, Macia L, Combe B, Morel J, Daien CI, Audo R. #CXCR5/CXCL13 pathway, a key driver for migration of regulatory B10 cells, is defective in patients with rheumatoid arthritis. Rheumatology (Oxford) 2021; 61:2185-2196. [PMID: 34382069 DOI: 10.1093/rheumatology/keab639] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/23/2021] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Chemokines (CKs) are key players of immune-cell homing and differentiation. CK receptors (CKRs) can be used to define T-cell functional subsets. We aimed to characterize the CKR profile of the regulatory B-cell subset B10+ cells and investigate the CKs involved in their migration and differentiation in healthy donors (CTLs) and patients with rheumatoid arthritis (RA). METHODS RNA sequencing and cytometry were used to compare CKR expression between B10+ and B10neg cells. Migration of B10+ and B10neg cells and interleukin 10 (IL-10) secretion of B cells in response to recombinant CKs or synovial fluid (SF) were assessed. RESULTS CXCR5 was expressed at a higher level on the B10+ cell surface as compared with other B cells (referred to as B10neg cells). In line with this, its ligand CXCL13 preferentially attracted B10+ cells over B10neg cells. Interestingly, synovial fluid from RA patients contained high levels of CXCL13 and induced strong and preferential migration of B10+ cells. Besides its role in attracting B10+ cells, CXCL13 also promoted IL-10 secretion by B cells. In RA patients, the level of CXCR5 on B cell surface was reduced. The preferential migration of RA B10+ cells toward CXCL13-rich SF was lost and CXCL13 stimulation triggered less IL-10 secretion than in healthy donors. CONCLUSION Our results identify that the CXCR5/CXCL13 axis is essential for B10+ cell biology but is defective in RA. Restoring the preferential migration of B10+ within the affected joints to better control inflammation may be part of therapeutic approach for RA.
Collapse
Affiliation(s)
- Claire Rempenault
- CHU and University of Montpellier, Rheumatology, Montpellier, France
| | - Julie Mielle
- IGMM, University of Montpellier, CNRS, Montpellier, France
| | | | - Pierre Corbeau
- CHU and University of Montpellier, Immunology, Nîmes, France.,IGH, CNRS, Montpellier, France (
| | - Laurence Macia
- Charles Perkins Centre, The University of Sydney, Sydney, Australia
| | - Bernard Combe
- CHU and University of Montpellier, Rheumatology, Montpellier, France
| | - Jacques Morel
- CHU and University of Montpellier, Rheumatology, Montpellier, France
| | - Claire Immediato Daien
- CHU and University of Montpellier, Rheumatology, Montpellier, France.,IGMM, University of Montpellier, CNRS, Montpellier, France
| | - Rachel Audo
- CHU and University of Montpellier, Rheumatology, Montpellier, France.,IGMM, University of Montpellier, CNRS, Montpellier, France
| |
Collapse
|
41
|
Smith LK, Fawaz K, Treanor B. Galectin-9 regulates the threshold of B cell activation and autoimmunity. eLife 2021; 10:64557. [PMID: 34369876 PMCID: PMC8352593 DOI: 10.7554/elife.64557] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 07/05/2021] [Indexed: 12/30/2022] Open
Abstract
Despite the mechanisms of central and peripheral tolerance, the mature B cell compartment contains cells reactive for self-antigen. How these cells are poised not to respond and the mechanisms that restrain B cell responses to low-affinity endogenous antigens are not fully understood. Here, we demonstrate a critical role for the glycan-binding protein galectin-9 in setting the threshold of B cell activation and that loss of this regulatory network is sufficient to drive spontaneous autoimmunity. We further demonstrate a critical role for galectin-9 in restraining not only conventional B-2 B cells, but also innate-like B-1a cells. We show that galectin-9-deficient mice have an expanded population of B-1a cells and increased titers of B-1a-derived autoantibodies. Mechanistically, we demonstrate that galectin-9 regulates BCR and distinct TLR responses in B-1a cells, but not B-1b cells, by regulating the interaction between BCR and TLRs with the regulatory molecules CD5 and CD180, respectively. In the absence of galectin-9, B-1a cells are more readily activated and secrete increased titers of autoantibodies that facilitate autoantigen delivery to the spleen, driving autoimmune responses.
Collapse
Affiliation(s)
- Logan K Smith
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Canada.,Department of Immunology, University of Toronto, Toronto, Canada
| | - Kareem Fawaz
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Canada
| | - Bebhinn Treanor
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Canada.,Department of Immunology, University of Toronto, Toronto, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| |
Collapse
|
42
|
Broadway R, Patel NM, Hillier LE, El-Briri A, Korneva YS, Zinovkin DA, Pranjol MZI. Potential Role of Diabetes Mellitus-Associated T Cell Senescence in Epithelial Ovarian Cancer Omental Metastasis. Life (Basel) 2021; 11:788. [PMID: 34440532 PMCID: PMC8401827 DOI: 10.3390/life11080788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 01/21/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is one of the most common causes of cancer-related deaths among women and is associated with age and age-related diseases. With increasing evidence of risks associated with metabolic inflammatory conditions, such as obesity and type 2 diabetes mellitus (T2DM), it is important to understand the complex pathophysiological mechanisms underlying cancer progression and metastasis. Age-related conditions can lead to both genotypic and phenotypic immune function alterations, such as induction of senescence, which can contribute to disease progression. Immune senescence is a common phenomenon in the ageing population, which is now known to play a role in multiple diseases, often detrimentally. EOC progression and metastasis, with the highest rates in the 75-79 age group in women, have been shown to be influenced by immune cells within the "milky spots" or immune clusters of the omentum. As T2DM has been reported to cause T cell senescence in both prediabetic and diabetic patients, there is a possibility that poor prognosis in EOC patients with T2DM is partly due to the accumulation of senescent T cells in the omentum. In this review, we explore this hypothesis with recent findings, potential therapeutic approaches, and future directions.
Collapse
Affiliation(s)
- Rhianne Broadway
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK; (R.B.); (L.E.H.)
| | - Nikita M. Patel
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, London EC1M 6BQ, UK; (N.M.P.); (A.E.-B.)
| | - Lucy E. Hillier
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK; (R.B.); (L.E.H.)
| | - Amal El-Briri
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, London EC1M 6BQ, UK; (N.M.P.); (A.E.-B.)
| | - Yulia S. Korneva
- Department of Pathological Anatomy, Smolensk State Medical University, Krupskoy St., 28, 214019 Smolensk, Russia;
- Smolensk Regional Institute of Pathology, Gagarina av, 214020 Smolensk, Russia
| | - Dmitry A. Zinovkin
- Department of Pathology, Gomel State Medical University, 246000 Gomel Region, Belarus;
| | - Md Zahidul I. Pranjol
- School of Life Sciences, University of Sussex, Falmer, Brighton BN1 9QG, UK; (R.B.); (L.E.H.)
| |
Collapse
|
43
|
Riese J, Gromann A, Lührs F, Kleinwort A, Schulze T. Sphingosine-1-Phosphate Receptor Type 4 (S1P 4) Is Differentially Regulated in Peritoneal B1 B Cells upon TLR4 Stimulation and Facilitates the Egress of Peritoneal B1a B Cells and Subsequent Accumulation of Splenic IRA B Cells under Inflammatory Conditions. Int J Mol Sci 2021; 22:ijms22073465. [PMID: 33801658 PMCID: PMC8037865 DOI: 10.3390/ijms22073465] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 02/08/2023] Open
Abstract
Background: Gram-negative infections of the peritoneal cavity result in profound modifications of peritoneal B cell populations and induce the migration of peritoneal B cells to distant secondary lymphoid organs. However, mechanisms controlling the egress of peritoneal B cells from the peritoneal cavity and their subsequent trafficking remain incompletely understood. Sphingosine-1-phosphate (S1P)-mediated signaling controls migratory processes in numerous immune cells. The present work investigates the role of S1P-mediated signaling in peritoneal B cell trafficking under inflammatory conditions. Methods: Differential S1P receptor expression after peritoneal B cell activation was assessed semi‑quantitatively using RT-PCR in vitro. The functional implications of differential S1P1 and S1P4 expression were assessed by transwell migration in vitro, by adoptive peritoneal B cell transfer in a model of sterile lipopolysaccharide (LPS)‑induced peritonitis and in the polymicrobial colon ascendens stent peritonitis (CASP) model. Results: The two sphingosine-1-phosphate receptors (S1PRs) expressed in peritoneal B cell subsets S1P1 and S1P4 are differentially regulated upon stimulation with the TLR4 agonist LPS, but not upon PMA/ionomycin or B cell receptor (BCR) crosslinking. S1P4 deficiency affects both the trafficking of activated peritoneal B cells to secondary lymphoid organs and the positioning of these cells within the functional compartments of the targeted organ. S1P4 deficiency in LPS-activated peritoneal B cells results in significantly reduced numbers of splenic innate response activator B cells. Conclusions: The S1P-S1PR system is implicated in the trafficking of LPS-activated peritoneal B cells. Given the protective role of peritoneal B1a B cells in peritoneal sepsis, further experiments to investigate the impact of S1P4-mediated signaling on the severity and mortality of peritoneal sepsis are warranted.
Collapse
Affiliation(s)
- Janik Riese
- Experimental Surgical Research Laboratory, Department of General Surgery, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, 17475 Greifswald, Germany
| | - Alina Gromann
- Experimental Surgical Research Laboratory, Department of General Surgery, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, 17475 Greifswald, Germany
| | - Felix Lührs
- Experimental Surgical Research Laboratory, Department of General Surgery, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, 17475 Greifswald, Germany
| | - Annabel Kleinwort
- Experimental Surgical Research Laboratory, Department of General Surgery, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, 17475 Greifswald, Germany
| | - Tobias Schulze
- Experimental Surgical Research Laboratory, Department of General Surgery, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, 17475 Greifswald, Germany
| |
Collapse
|
44
|
Innate Lymphoid Cells Play a Pathogenic Role in Pericarditis. Cell Rep 2021; 30:2989-3003.e6. [PMID: 32130902 PMCID: PMC7332109 DOI: 10.1016/j.celrep.2020.02.040] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/05/2019] [Accepted: 02/07/2020] [Indexed: 12/27/2022] Open
Abstract
We find that cardiac group 2 innate lymphoid cells (ILC2s) are essential for the development of IL-33-induced eosinophilic pericarditis. We show a pathogenic role for ILC2s in cardiac inflammation, in which ILC2s activated by IL-33 drive the development of eosinophilic pericarditis in collaboration with cardiac fibroblasts. ILCs, not T and B cells, are required for the development of pericarditis. ILC2s transferred to the heart of Rag2-/-Il2rg-/- mice restore their susceptibility to eosinophil infiltration. Moreover, ILC2s direct cardiac fibroblasts to produce eotaxin-1. We also find that eosinophils reside in the mediastinal cavity and that eosinophils transferred to the mediastinal cavity of eosinophil-deficient ΔdblGATA1 mice following IL-33 treatment migrate to the heart. Thus, the serous cavities may serve as a reservoir of cardiac-infiltrating eosinophils. In humans, patients with pericarditis show higher amounts of ILCs in pericardial fluid than do healthy controls and patients with other cardiac diseases. We demonstrate that ILCs play a critical role in pericarditis.
Collapse
|
45
|
Louwe PA, Badiola Gomez L, Webster H, Perona-Wright G, Bain CC, Forbes SJ, Jenkins SJ. Recruited macrophages that colonize the post-inflammatory peritoneal niche convert into functionally divergent resident cells. Nat Commun 2021; 12:1770. [PMID: 33741914 PMCID: PMC7979918 DOI: 10.1038/s41467-021-21778-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammation generally leads to recruitment of monocyte-derived macrophages. What regulates the fate of these cells and to what extent they can assume the identity and function of resident macrophages is unclear. Here, we show that macrophages elicited into the peritoneal cavity during mild inflammation persist long-term but are retained in an immature transitory state of differentiation due to the presence of enduring resident macrophages. By contrast, severe inflammation results in ablation of resident macrophages and a protracted phase wherein the cavity is incapable of sustaining a resident phenotype, yet ultimately elicited cells acquire a mature resident identity. These macrophages also have transcriptionally and functionally divergent features that result from inflammation-driven alterations to the peritoneal cavity micro-environment and, to a lesser extent, effects of origin and time-of-residency. Hence, rather than being predetermined, the fate of inflammation-elicited peritoneal macrophages seems to be regulated by the environment.
Collapse
Affiliation(s)
- P A Louwe
- Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh, EH16 4TJ, United Kingdom
| | - L Badiola Gomez
- Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh, EH16 4TJ, United Kingdom
| | - H Webster
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - G Perona-Wright
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - C C Bain
- Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh, EH16 4TJ, United Kingdom
| | - S J Forbes
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - S J Jenkins
- Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh, EH16 4TJ, United Kingdom.
| |
Collapse
|
46
|
Abstract
B cell subsets differ in development, tissue distribution, and mechanisms of activation. In response to infections, however, all can differentiate into extrafollicular plasmablasts that rapidly provide highly protective antibodies, indicating that these plasmablasts are the main humoral immune response effectors. Yet, the effectiveness of this response type depends on the presence of antigen-specific precursors in the circulating mature B cell pool, a pool that is generated initially through the stochastic processes of B cell receptor assembly. Importantly, germinal centers then mold the repertoire of this B cell pool to be increasingly responsive to pathogens by generating a broad array of antimicrobial memory B cells that act as highly effective precursors of extrafollicular plasmablasts. Such B cell repertoire molding occurs in two ways: continuously via the chronic germinal centers of mucosal lymphoid tissues, driven by the presence of the microbiome, and via de novo generated germinal centers following acute infections. For effectively evaluating humoral immunity as a correlate of immune protection, it might be critical to measure memory B cell pools in addition to antibody titers.
Collapse
Affiliation(s)
- Nicole Baumgarth
- Center for Immunology and Infectious Diseases and Department of Pathology, Microbiology and Immunology, University of California, Davis, California 95616, USA;
| |
Collapse
|
47
|
Abstract
B-1 cells are fetal-origin B lymphocytes with unique developmental and functional characteristics that can generate natural, polyreactive antibodies with important functions in tissue homeostasis and immune defense. While B-1 cell frequencies in bone marrow and secondary lymphoid tissues are low, relative high frequencies exist within peritoneal and pleural cavities of mice, including both CD5+ and CD5- B-1 cells. These cells represent B-1 reservoirs that, when activated, migrate to lymphoid tissues to secrete antibodies and/or cytokines. Here, we outline efficient methods for the extraction and magnetic isolation of CD5+ B-1 cells from the peritoneal and pleural cavities as well as the separation and phenotypic characterization of CD5+ and CD5- B-1 cells by flow cytometry.
Collapse
|
48
|
Jaufmann J, Carevic M, Tümen L, Eliacik D, Schmitt F, Hartl D, Beer-Hammer S. Enhanced IgG 1 -mediated antibody response towards thymus-dependent immunization in CXCR1-deficient mice. IMMUNITY INFLAMMATION AND DISEASE 2020; 9:210-222. [PMID: 33226189 PMCID: PMC7860589 DOI: 10.1002/iid3.380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023]
Abstract
Background Chemokine receptors and their corresponding ligands are key players of immunity by regulation of immune cell differentiation and migration. CXCR1 is a high‐affinity receptor for CXCL8. Differential expression of CXCR1 is associated with a variety of human pathologies including cancer and inflammatory diseases. While various studies have highlighted the importance of CXCR1‐mediated CXCL8‐sensing for neutrophil trafficking and function, its role in B‐cell responses remains unsolved. Therefore, our aim was to investigate innate and adaptive antibody responses in CXCR1‐deficient mice. Methods Cell populations of the spleen and the peritoneal cavity were identified and quantified via flow cytometry. To investigate thymus‐independent (TI) and thymus‐dependent (TD) antibody responses, mice were immunized intraperitoneally with TNP‐Ficoll, Pneumovax23, and TNP‐Chicken Gamma Globulin. Mice were bled before as well as 7 and 14 days after vaccination to collect serum. Serum antibody levels overtime were analyzed according to their specificity by enzyme‐linked immunosorbent assay. B‐1 cell functionality was examined by IL‐5/IL‐5Rα‐dependent stimulation of peritoneal and splenic cells in vitro. To analyze CXCR1/2‐expression, CD19+ splenocytes were enriched by magnetic‐activated cell sorting before isolation of total RNA contents, followed by reverse transcription and real‐time polymerase chain reaction. Results The distribution of natural B‐1 cell populations was disturbed in the absence of CXCR1, while their responsiveness towards TI antigens and in vitro stimulation remained functional. Besides, CXCR1‐deficiency was accompanied by increased frequencies of follicular B‐2 cells in the spleen. Interestingly, these mice produced elevated levels of antigen‐specific IgG1 upon TD immunization and harbored a significantly enlarged proportion of CXCR5‐expressing T helper (H) cells. CXCR1‐expression was detectable in CD19+ splenocytes derived from wild‐type, but not CXCR1‐deficient mice. Conclusion Our data demonstrate a previously unknown relevance of CXCR1 for the production of specific IgG1 in response to vaccination. These findings identify CXCR1 as a promising candidate for future studies on the regulation of adaptive antibody responses.
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
| | - Melanie Carevic
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, 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
| | - Derya Eliacik
- Department of Pharmacology, Experimental Therapy, and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Fee Schmitt
- Department of Pharmacology, Experimental Therapy, and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Dominik Hartl
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tuebingen, Tuebingen, Germany.,Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - 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
|
49
|
Jia X, Berta G, Gábris F, Kellermayer Z, Balogh P. Role of adipose-associated lymphoid tissues in the immunological homeostasis of the serosal surface. Immunol Lett 2020; 228:135-141. [PMID: 33166529 DOI: 10.1016/j.imlet.2020.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/13/2020] [Accepted: 11/03/2020] [Indexed: 11/30/2022]
Abstract
Although not typical lymphoid organs, analysis of the visceral adipose-associated lymphoid tissues has recently substantially expanded our knowledge about the immunological features of these elusive compartments. Recent data have highlighted their considerable complexity in cellular organization and interactions in several biological processes, including adaptive immune responses, tissue plasticity to accommodate mesenchymal stem cells and progenitors, and providing a suitable microenvironment for serosal tumor propagation. This review aims to present a comprehensive view of the adipose-associated lymphoid tissues in local and systemic immune responsiveness, with particular emphasis on the omental and mesenteric lymphoid tissues in the serosal defense of abdominal organs.
Collapse
Affiliation(s)
- Xinkai Jia
- Department of Immunology and Biotechnology, Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Hungary
| | - Fanni Gábris
- Department of Immunology and Biotechnology, Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary
| | - Zoltán Kellermayer
- Department of Immunology and Biotechnology, Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary
| | - Péter Balogh
- Department of Immunology and Biotechnology, Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary; Lymphoid Organogenesis Research Group, Szentágothai Research Center, University of Pécs Medical School, Hungary.
| |
Collapse
|
50
|
Jaufmann J, Tümen L, Schmitt F, Schäll D, von Holleben M, Beer-Hammer S. SLy2-deficiency promotes B-1 cell immunity and triggers enhanced production of IgM and IgG 2 antibodies against pneumococcal vaccine. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:736-752. [PMID: 33098380 PMCID: PMC7654406 DOI: 10.1002/iid3.365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/15/2020] [Accepted: 10/13/2020] [Indexed: 01/01/2023]
Abstract
Background Despite the benefits of existing vaccines, Streptococcus pneumoniae is still responsible for the greatest proportion of respiratory tract infections around the globe, thereby substantially contributing to morbidity and mortality in humans. B‐1 cells are key players of bacterial clearance during pneumococcal infection and even provide long‐lasting immunity towards S. pneumoniae. Previous reports strongly suggest an essential role of the immunoinhibitory adapter Src homology domain 3 lymphocyte protein 2 (SLy2) for B‐1 cell‐mediated antibody production. The objective of this study is to evaluate S. pneumoniae‐directed B cell responses in the context of SLy2 deficiency. Methods B‐1 cell populations were analyzed via flow cytometry before and after pneumococcal immunization of SLy2‐deficient and wild‐type control mice. Global and vaccine‐specific immunoglobulin M (IgM) and IgG antibody titers were assessed by enzyme‐linked immunosorbent assay. To investigate survival rates during acute pneumococcal lung infection, mice were intranasally challenged with S. pneumoniae (serotype 3). Complementary isolated splenic B cells were stimulated in vitro and their proliferative response was assessed by fluorescent staining. In vitro antibody secretion was quantified by LEGENDplex. Results We demonstrate increased frequencies of B‐1 cells and elevated titers of preantigenic IgM in SLy2‐deficient mice. In addition, these mice produce significantly more amounts of IgM and IgG2 upon pneumococcal vaccination. Knocking out SLy2 did not induce survival advantages in our murine model of acute pneumonia, indicating the presence of compensatory mechanisms. Conclusion Our results reveal reinforced specific antibody responses towards pneumococcal polysaccharides and enhanced IgG2 secretion as a consequence of SLy2 deficiency, which could be relevant to the development of more efficient vaccines.
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
| | - Fee Schmitt
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Daniel Schäll
- Department of Pharmacology, Experimental Therapy and Toxicology, Institute of Experimental and Clinical Pharmacology and Pharmacogenomik and ICePhA, University of Tuebingen, Tuebingen, Germany
| | - Max von Holleben
- Institute for Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University, Duesseldorf, 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.,Institute for Medical Microbiology and Hospital Hygiene, Heinrich-Heine-University, Duesseldorf, Germany
| |
Collapse
|