1
|
Wang Y, Fei Y. Causal relationship between 731 immune cells and the risk of myeloproliferative neoplasms: A 2-sample bidirectional Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40945. [PMID: 39705412 PMCID: PMC11666147 DOI: 10.1097/md.0000000000040945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/03/2024] [Accepted: 11/25/2024] [Indexed: 12/22/2024] Open
Abstract
Myeloproliferative neoplasms (MPN) are chronic hematological disorders marked by the abnormal proliferation of bone marrow cells. The most commonly encountered forms are polycythemia vera (PV), primary myelofibrosis (PMF), and essential thrombocythemia (ET). These disorders are generally associated with increases in blood components, which can lead to conditions like splenomegaly, thrombosis, bleeding tendencies, and a heightened risk of progressing to acute leukemia. Previous research has indicated a possible link between immune cells and MPN, yet this association is still poorly understood. This study seeks to elucidate the causal relationship between immune cell characteristics and the development of MPN. In this study, we employed Mendelian randomization (MR) to investigate potential causal links between 731 immune cell traits and the risk of developing MPN, leveraging data from genome-wide association studies (GWAS). To ensure the robustness of our findings, we conducted extensive sensitivity analyses to assess heterogeneity and detect any pleiotropic effects. Moreover, we implemented a false discovery rate (FDR) correction to mitigate the risk of false positives that may result from the multiple hypothesis testing, thereby adjusting for any statistical biases due to multiple comparisons. The immune phenotype IgD on IgD+ CD24- B cells demonstrated a statistically significant protective effect against MPN (PFDR = 0.047). Upon adjusting the significance threshold to PFDR < 0.20, 16 immune cell phenotypes were significantly associated with MPN. Among these, 11 were found to exert a protective effect against MPN, 5 phenotypes were associated with an elevated risk of MPN. This research highlights a significant association between various immune cell phenotypes and the risk of developing MPN, thereby advancing our understanding of the intricate interplay between immune cell traits and the progression of MPN.
Collapse
Affiliation(s)
- Yao Wang
- Department of Trauma and Orthopaedic, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Yang Fei
- Department of Hematology and Oncology, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
2
|
Liu JC, Zeng Q, Duan YG, Yeung WSB, Li RHW, Ng EHY, Cheung KW, Zhang Q, Chiu PCN. B cells: roles in physiology and pathology of pregnancy. Front Immunol 2024; 15:1456171. [PMID: 39434884 PMCID: PMC11491347 DOI: 10.3389/fimmu.2024.1456171] [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: 06/28/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
B cells constitute a diverse and adaptable immune cell population with functions that can vary according to the environment and circumstances. The involvement of B cells in pregnancy, as well as the associated molecular pathways, has yet to be investigated. This review consolidates current knowledge on B cell activities and regulation during pregnancy, with a particular focus on the roles of various B cell subsets and the effects of B cell-derived factors on pregnancy outcomes. Moreover, the review examines the significance of B cell-associated autoantibodies, cytokines, and signaling pathways in relation to pregnancy complications such as pregnancy loss, preeclampsia, and preterm birth.
Collapse
Affiliation(s)
- Jin-Chuan Liu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qunxiong Zeng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong-Gang Duan
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - William S. B. Yeung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Raymond H. W. Li
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ernest H. Y. Ng
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ka-Wang Cheung
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Philip C. N. Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing (LKS) Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
3
|
Gao H, Zhao Y, Zhao S, Dai XQ, Qin XY, Zheng WL, He TT, Zhang N, Zhu C, Wang HM, Pan W, Zhu XM, Gao XM, Dai JF, Gong FY, Wang J. The ICF2 gene Zbtb24 specifically regulates the differentiation of B1 cells via promoting heme synthesis. Cell Mol Biol Lett 2024; 29:123. [PMID: 39277732 PMCID: PMC11401330 DOI: 10.1186/s11658-024-00641-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Loss-of-function mutations of ZBTB24 cause immunodeficiency, centromeric instability, and facial anomalies syndrome 2 (ICF2). ICF2 is a rare autosomal recessive disorder with immunological defects in serum antibodies and circulating memory B cells, resulting in recurrent and sometimes fatal respiratory and gastrointestinal infections. The genotype-phenotype correlation in patients with ICF2 indicates an essential role of ZBTB24 in the terminal differentiation of B cells. METHODS We used the clustered regularly interspaced short palindromic repeats (CRISPER)/Cas9 technology to generate B cell specific Zbtb24-deficient mice and verified the deletion specificity and efficiency by quantitative polymerase chain reaction (Q-PCR) and western blotting analyses in fluorescence-activated cell sorting (FACS)-sorted cells. The development, phenotype of B cells and in vivo responses to T cell dependent or independent antigens post immunization were analyzed by flow cytometry and enzyme-linked immunosorbent assay (ELISA). Adoptive transfer experiment in combination with in vitro cultures of FACS-purified B cells and RNA-Seq analysis were utilized to specifically determine the impact of Zbtb24 on B cell biology as well as the underlying mechanisms. RESULTS Zbtb24 is dispensable for B cell development and maintenance in naive mice. Surprisingly, B cell specific deletion of Zbtb24 does not evidently compromise germinal center reactions and the resulting primary and secondary antibody responses induced by T cell dependent antigens (TD-Ags), but significantly inhibits T cell independent antigen-elicited antibody productions in vivo. At the cellular level, Zbtb24-deficiency specifically impedes the plasma cell differentiation of B1 cells without impairing their survival, activation and proliferation in vitro. Mechanistically, Zbtb24-ablation attenuates heme biosynthesis partially through mTORC1 in B1 cells, and addition of exogenous hemin abrogates the differentiation defects of Zbtb24-null B1 cells. CONCLUSIONS Zbtb24 seems to regulate antibody responses against TD-Ags B cell extrinsically, but it specifically promotes the plasma cell differentiation of B1 cells via heme synthesis in mice. Our study also suggests that defected B1 functions contribute to recurrent infections in patients with ICF2.
Collapse
Affiliation(s)
- He Gao
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Ying Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Sai Zhao
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiao-Qiu Dai
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiao-Yuan Qin
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Wei-Long Zheng
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Ting-Ting He
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Nan Zhang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Can Zhu
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Hong-Min Wang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Wen Pan
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xue-Mei Zhu
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiao-Ming Gao
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Jian-Feng Dai
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Fang-Yuan Gong
- Department of Immunology, School of Basic Medical Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Jun Wang
- Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
4
|
Murata K, Murao A, Tan C, Wang P, Aziz M. B-1a cells scavenge NETs to attenuate sepsis. J Leukoc Biol 2024; 116:632-643. [PMID: 38484156 PMCID: PMC11367732 DOI: 10.1093/jleuko/qiae066] [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: 08/16/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 09/03/2024] Open
Abstract
B-1a cells, a regulatory subset of B lymphocytes, produce natural IgM and interleukin-10. Neutrophil extracellular traps (NETs) play a crucial role in pathogen defense, but their excessive formation during sepsis can cause further inflammation and tissue damage. In sepsis, extracellular cold-inducible RNA-binding protein (eCIRP), a damage-associated molecular pattern, is released to induce NET formation. We hypothesize that B-1a cells clear NETs to prevent sepsis-induced injury. Sepsis in mice was induced by injecting 1 × 107 and 5 × 107 colony-forming units of Escherichia coli intraperitoneally. After 4 and 20 h, we assessed the number of B-1a cells in the peritoneal cavity using flow cytometry. Our results showed that the number of peritoneal B-1a cells was significantly decreased in E. coli sepsis mice. Importantly, replenishing B-1a cells via intraperitoneal injection in sepsis mice significantly decreased NETs in peritoneal neutrophils. We also observed a decrease in serum inflammation and injury markers and a significant increase in the overall survival rate in B-1a cell-treated septic mice. To understand the mechanism, we cocultured bone marrow-derived neutrophils with peritoneal B-1a cells in a contact or noncontact condition using an insert and stimulated them with eCIRP. After 4 h, we found that eCIRP significantly increased NET formation in bone marrow-derived neutrophils. Interestingly, we observed that B-1a cells inhibited NETs by 67% in a contact-dependent manner. Surprisingly, when B-1a cells were cultured in inserts, there was no significant decrease in NET formation, suggesting that direct cell-to-cell contact is crucial for this inhibitory effect. We further determined that B-1a cells promoted NET phagocytosis, and this was mediated through natural IgM, as blocking the IgM receptor attenuated the engulfment of NETs by B-1a cells. Finally, we identified that following their engulfment, NETs were localized into the lysosomal compartment for lysis. Thus, our study suggests that B-1a cells decrease NET content in eCIRP-treated neutrophils and E. coli sepsis mice.
Collapse
Affiliation(s)
- Kensuke Murata
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Chuyi Tan
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Ping Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
| |
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
|
Ma Y, Lai J, Wan Q, Chen Z, Sun L, Zhang Q, Guan C, Li Q, Wu J. Identification of common mechanisms and biomarkers for dermatomyositis and atherosclerosis based on bioinformatics analysis. Skin Res Technol 2024; 30:e13808. [PMID: 38899746 PMCID: PMC11187814 DOI: 10.1111/srt.13808] [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: 04/13/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Dermatomyositis (DM) manifests as an autoimmune and inflammatory condition, clinically characterized by subacute progressive proximal muscle weakness, rashes or both along with extramuscular manifestations. Literature indicates that DM shares common risk factors with atherosclerosis (AS), and they often co-occur, yet the etiology and pathogenesis remain to be fully elucidated. This investigation aims to utilize bioinformatics methods to clarify the crucial genes and pathways that influence the pathophysiology of both DM and AS. METHOD Microarray datasets for DM (GSE128470, GSE1551, GSE143323) and AS (GSE100927, GSE28829, GSE43292) were retrieved from the Gene Expression Omnibus (GEO) database. The weighted gene co-expression network analysis (WGCNA) was used to reveal their co-expressed modules. Differentially expression genes (DEGs) were identified using the "limma" package in R software, and the functions of common DEGs were determined by functional enrichment analysis. A protein-protein interaction (PPI) network was established using the STRING database, with central genes evaluated by the cytoHubba plugin, and validated through external datasets. Immune infiltration analysis of the hub genes was conducted using the CIBERSORT method, along with Gene Set Enrichment Analysis (GSEA). Finally, the NetworkAnalyst platform was employed to examine the transcription factors (TFs) responsible for regulating pivotal crosstalk genes. RESULTS Utilizing WGCNA analysis, a total of 271 overlapping genes were pinpointed. Subsequent DEG analysis revealed 34 genes that are commonly found in both DM and AS, including 31 upregulated genes and 3 downregulated genes. The Degree Centrality algorithm was applied separately to the WGCNA and DEG collections to select the 15 genes with the highest connectivity, and crossing the two gene sets yielded 3 hub genes (PTPRC, TYROBP, CXCR4). Validation with external datasets showed their diagnostic value for DM and AS. Analysis of immune infiltration indicates that lymphocytes and macrophages are significantly associated with the pathogenesis of DM and AS. Moreover, GSEA analysis suggested that the shared genes are enriched in various receptor interactions and multiple cytokines and receptor signaling pathways. We coupled the 3 hub genes with their respective predicted genes, identifying a potential key TF, CBFB, which interacts with all 3 hub genes. CONCLUSION This research utilized comprehensive bioinformatics techniques to explore the shared pathogenesis of DM and AS. The three key genes, including PTPRC, TYROBP, and CXCR4, are related to the pathogenesis of DM and AS. The central genes and their correlations with immune cells may serve as potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Yirong Ma
- Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Junyu Lai
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Qiang Wan
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Zhengtao Chen
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Liqiang Sun
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Qinhe Zhang
- Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Chengyan Guan
- Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Qiming Li
- Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| | - Jianguang Wu
- Department of cardiovascularAffiliated Hospital of Jiangxi University of Traditional Chinese MedicineNanchangJiangxiChina
| |
Collapse
|
7
|
Obare LM, Bonami RH, Doran A, Wanjalla CN. B cells and atherosclerosis: A HIV perspective. J Cell Physiol 2024; 239:e31270. [PMID: 38651687 PMCID: PMC11209796 DOI: 10.1002/jcp.31270] [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] [Revised: 03/09/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
Atherosclerosis remains a leading cause of cardiovascular disease (CVD) globally, with the complex interplay of inflammation and lipid metabolism at its core. Recent evidence suggests a role of B cells in the pathogenesis of atherosclerosis; however, this relationship remains poorly understood, particularly in the context of HIV. We review the multifaceted functions of B cells in atherosclerosis, with a specific focus on HIV. Unique to atherosclerosis is the pivotal role of natural antibodies, particularly those targeting oxidized epitopes abundant in modified lipoproteins and cellular debris. B cells can exert control over cellular immune responses within atherosclerotic arteries through antigen presentation, chemokine production, cytokine production, and cell-cell interactions, actively participating in local and systemic immune responses. We explore how HIV, characterized by chronic immune activation and dysregulation, influences B cells in the context of atherosclerosis, potentially exacerbating CVD risk in persons with HIV. By examining the proatherogenic and antiatherogenic properties of B cells, we aim to deepen our understanding of how B cells influence atherosclerotic plaque development, especially within the framework of HIV. This research provides a foundation for novel B cell-targeted interventions, with the potential to mitigate inflammation-driven cardiovascular events, offering new perspectives on CVD risk management in PLWH.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel H. Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amanda Doran
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
8
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
9
|
Santacroce E, D’Angerio M, Ciobanu AL, Masini L, Lo Tartaro D, Coloretti I, Busani S, Rubio I, Meschiari M, Franceschini E, Mussini C, Girardis M, Gibellini L, Cossarizza A, De Biasi S. Advances and Challenges in Sepsis Management: Modern Tools and Future Directions. Cells 2024; 13:439. [PMID: 38474403 PMCID: PMC10931424 DOI: 10.3390/cells13050439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Sepsis, a critical condition marked by systemic inflammation, profoundly impacts both innate and adaptive immunity, often resulting in lymphopenia. This immune alteration can spare regulatory T cells (Tregs) but significantly affects other lymphocyte subsets, leading to diminished effector functions, altered cytokine profiles, and metabolic changes. The complexity of sepsis stems not only from its pathophysiology but also from the heterogeneity of patient responses, posing significant challenges in developing universally effective therapies. This review emphasizes the importance of phenotyping in sepsis to enhance patient-specific diagnostic and therapeutic strategies. Phenotyping immune cells, which categorizes patients based on clinical and immunological characteristics, is pivotal for tailoring treatment approaches. Flow cytometry emerges as a crucial tool in this endeavor, offering rapid, low cost and detailed analysis of immune cell populations and their functional states. Indeed, this technology facilitates the understanding of immune dysfunctions in sepsis and contributes to the identification of novel biomarkers. Our review underscores the potential of integrating flow cytometry with omics data, machine learning and clinical observations to refine sepsis management, highlighting the shift towards personalized medicine in critical care. This approach could lead to more precise interventions, improving outcomes in this heterogeneously affected patient population.
Collapse
Affiliation(s)
- Elena Santacroce
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Miriam D’Angerio
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Alin Liviu Ciobanu
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Linda Masini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Irene Coloretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care, Jena University Hospital, 07747 Jena, Germany;
| | - Marianna Meschiari
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Erica Franceschini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Cristina Mussini
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, 41121 Modena, Italy; (I.C.); (S.B.); (M.M.); (E.F.); (C.M.); (M.G.)
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy; (E.S.); (M.D.); (A.L.C.); (L.M.); (D.L.T.); (L.G.); (A.C.)
| |
Collapse
|
10
|
Fan S, Kang B, Li S, Li W, Chen C, Chen J, Deng L, Chen D, Zhou J. Exploring the multifaceted role of RASGRP1 in disease: immune, neural, metabolic, and oncogenic perspectives. Cell Cycle 2024; 23:722-746. [PMID: 38865342 PMCID: PMC11229727 DOI: 10.1080/15384101.2024.2366009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/25/2023] [Indexed: 06/14/2024] Open
Abstract
RAS guanyl releasing protein 1 (RASGRP1) is a guanine nucleotide exchange factor (GEF) characterized by the presence of a RAS superfamily GEF domain. It functions as a diacylglycerol (DAG)-regulated nucleotide exchange factor, specifically activating RAS through the exchange of bound GDP for GTP. Activation of RAS by RASGRP1 has a wide range of downstream effects at the cellular level. Thus, it is not surprising that many diseases are associated with RASGRP1 disorders. Here, we present an overview of the structure and function of RASGRP1, its crucial role in the development, expression, and regulation of immune cells, and its involvement in various signaling pathways. This review comprehensively explores the relationship between RASGRP1 and various diseases, elucidates the underlying molecular mechanisms of RASGRP1 in each disease, and identifies potential therapeutic targets. This study provides novel insights into the role of RASGRP1 in insulin secretion and highlights its potential as a therapeutic target for diabetes. The limitations and challenges associated with studying RASGRP1 in disease are also discussed.
Collapse
Affiliation(s)
- Shangzhi Fan
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Kang
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shaoqian Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weiyi Li
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Canyu Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jixiang Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lijing Deng
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Danjun Chen
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiecan Zhou
- The First Affiliated Hospital, Hunan Provincial Clinical Medical Research Center for Drug Evaluation of Major Chronic Diseases,Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
11
|
Wang X, Wu H, Fang C, Li Z. Insights into innate immune cell evasion by Chlamydia trachomatis. Front Immunol 2024; 15:1289644. [PMID: 38333214 PMCID: PMC10850350 DOI: 10.3389/fimmu.2024.1289644] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Chlamydia trachomatis, is a kind of obligate intracellular pathogen. The removal of C. trachomatis relies primarily on specific cellular immunity. It is currently considered that CD4+ Th1 cytokine responses are the major protective immunity against C. trachomatis infection and reinfection rather than CD8+ T cells. The non-specific immunity (innate immunity) also plays an important role in the infection process. To survive inside the cells, the first process that C. trachomatis faces is the innate immune response. As the "sentry" of the body, mast cells attempt to engulf and remove C. trachomatis. Dendritic cells present antigen of C. trachomatis to the "commanders" (T cells) through MHC-I and MHC-II. IFN-γ produced by activated T cells and natural killer cells (NK) further activates macrophages. They form the body's "combat troops" and produce immunity against C. trachomatis in the tissues and blood. In addition, the role of eosinophils, basophils, innate lymphoid cells (ILCs), natural killer T (NKT) cells, γδT cells and B-1 cells should not be underestimated in the infection of C. trachomatis. The protective role of innate immunity is insufficient, and sexually transmitted diseases (STDs) caused by C. trachomatis infections tend to be insidious and recalcitrant. As a consequence, C. trachomatis has developed a unique evasion mechanism that triggers inflammatory immunopathology and acts as a bridge to protective to pathological adaptive immunity. This review focuses on the recent advances in how C. trachomatis evades various innate immune cells, which contributes to vaccine development and our understanding of the pathophysiologic consequences of C. trachomatis infection.
Collapse
Affiliation(s)
| | | | | | - Zhongyu Li
- Institute of Pathogenic Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, School of Nursing, Hengyang Medical College, University of South China, Hengyang, China
| |
Collapse
|
12
|
Najimi N, Kadi C, Elmtili N, Seghrouchni F, Bakri Y. Unravelling humoral immunity in SARS-CoV-2: Insights from infection and vaccination. Hum Antibodies 2024; 32:85-106. [PMID: 38758995 DOI: 10.3233/hab-230017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2024]
Abstract
Following infection and vaccination against SARS-CoV-2, humoral components of the adaptive immune system play a key role in protecting the host. Specifically, B cells generate high-affinity antibodies against various antigens of the virus. In this review, we discuss the mechanisms of immunity initiation through both natural infection and vaccination, shedding light on the activation of B cell subsets in response to SARS-CoV-2 infection and vaccination. The innate immune system serves as the initial line of primary and nonspecific defence against viruses. However, within several days following infection or a vaccine dose, a virus-specific immune response is initiated, primarily by B cells that produce antibodies. These antibodies contribute to the resolution of the disease. Subsequently, these B cells transition into memory B cells, which play a crucial role in providing long-term immunity against the virus. CD4+ T helper cells initiate a cascade, leading to B cell somatic hypermutation, germinal center memory B cells, and the production of neutralizing antibodies. B-cell dysfunction can worsen disease severity and reduce vaccine efficacy. Notably, individuals with B cell immunodeficiency show lower IL-6 production. Furthermore, this review delves into several aspects of immune responses, such as hybrid immunity, which has shown promise in boosting broad-spectrum protection. Cross-reactive immunity is under scrutiny as well, as pre-existing antibodies can offer protection against the disease. We also decipher breakthrough infection mechanisms, especially with the novel variants of the virus. Finally, we discuss some potential therapeutic solutions regarding B cells including convalescent plasma therapy, B-1 cells, B regulatory cell (Breg) modulation, and the use of neutralizing monoclonal antibodies in combating the infection. Ongoing research is crucial to grasp population immunity trends and assess the potential need for booster doses in maintaining effective immune responses against potential viral threats.
Collapse
Affiliation(s)
- Nouhaila Najimi
- Laboratory of Human Pathologies Biology and Center of Genomic of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Chaimae Kadi
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
- Laboratory of Biology and Health, Faculty of Sciences of Tétouan, Abdelmalek Essaâdi University, Tétouan, Morocco
| | - Noureddine Elmtili
- Laboratory of Biology and Health, Faculty of Sciences of Tétouan, Abdelmalek Essaâdi University, Tétouan, Morocco
| | - Fouad Seghrouchni
- Mohammed VI Center for Research and Innovation, Rabat, Morocco
- Mohammed VI University of Sciences and Health, Casablanca, Morocco
| | - Youssef Bakri
- Laboratory of Human Pathologies Biology and Center of Genomic of Human Pathologies Biology, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| |
Collapse
|
13
|
Kim CS, Jung MH, Choi EY, Shin DM. Probiotic supplementation has sex-dependent effects on immune responses in association with the gut microbiota in community-dwelling older adults: a randomized, double-blind, placebo-controlled, multicenter trial. Nutr Res Pract 2023; 17:883-898. [PMID: 37780220 PMCID: PMC10522805 DOI: 10.4162/nrp.2023.17.5.883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 05/13/2023] [Accepted: 05/19/2023] [Indexed: 10/03/2023] Open
Abstract
BACKGROUND/OBJECTIVES Probiotics have been suggested as potent modulators of age-related disorders in immunological functions, yet little is known about sex-dependent effects of probiotic supplements. Therefore, we aimed to investigate sex-dependent effects of probiotics on profiles of the gut microbiota and peripheral immune cells in healthy older adults. SUBJECTS/METHODS In a randomized, double-blind, placebo-controlled, multicenter trial, healthy elderly individuals ≥ 65 yrs old were administered probiotic capsules (or placebo) for 12 wk. Gut microbiota was analyzed using 16S rRNA gene sequencing and bioinformatic analyses. Peripheral immune cells were profiled using flow cytometry for lymphocytes (natural killer, B, CD4+ T, and CD8+ T cells), dendritic cells, monocytes, and their subpopulations. RESULTS Compared with placebo, phylum Firmicutes was significantly reduced in the probiotic group in women, but not in men. At the genus level, sex-specific responses included reductions in the relative abundances of pro-inflammatory gut microbes, including Catabacter and unclassified_Coriobacteriales, and Burkholderia and unclassified Enterobacteriaceae, in men and women, respectively. Peripheral immune cell profiling analysis revealed that in men, probiotics significantly reduced the proportions of dendritic cells and CD14+ CD16- monocytes; however, these effects were not observed in women. In contrast, the proportion of total CD4+ T cells was significantly reduced in women in the probiotic group. Additionally, serum lipopolysaccharide-binding protein levels showed a decreasing tendency that were positively associated with changes in gut bacteria, including Catabacter (ρ = 0.678, P < 0.05) and Burkholderia (ρ = 0.673, P < 0.05) in men and women, respectively. CONCLUSIONS These results suggest that probiotic supplementation may reduce the incidence of inflammation-related diseases by regulating the profiles of the gut microbiota and peripheral immune cells in healthy elders in a sex-specific manner.
Collapse
Affiliation(s)
- Chong-Su Kim
- Department of Food and Nutrition, College of Natural Information Sciences, Dongduk Women's University, Seoul 02748, Korea
| | - Min Ho Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
14
|
Kang M, Yadav MK, Mbanefo EC, Yu CR, Egwuagu CE. IL-27-containing exosomes secreted by innate B-1a cells suppress and ameliorate uveitis. Front Immunol 2023; 14:1071162. [PMID: 37334383 PMCID: PMC10272713 DOI: 10.3389/fimmu.2023.1071162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 05/18/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction IL-27 is a heterodimeric cytokine composed of Ebi3 and IL-27p28 and can exert proinflammatory or immune suppressive effects depending on the physiological context. Ebi3 does not contain membrane-anchoring motifs, suggesting that it is a secreted protein while IL-27p28 is poorly secreted. How IL-27p28 and Ebi3 dimerize in-vivo to form biologically active IL-27 is unknown. Major impediment to clinical use of IL-27 derives from difficulty of determining exact amount of bioavailable heterodimeric IL-27 needed for therapy. Methods To understand how IL-27 mediates immune suppression, we characterized an innate IL-27-producing B-1a regulatory B cell population (i27-Breg) and mechanisms i27-Bregs utilize to suppress neuroinflammation in mouse model of uveitis. We also investigated biosynthesis of IL-27 and i27-Breg immunobiology by FACS, immunohistochemical and confocal microscopy. Results Contrary to prevailing view that IL-27 is a soluble cytokine, we show that i27-Bregs express membrane-bound IL-27. Immunohistochemical and confocal analyses co-localized expression of IL-27p28 at the plasma membrane in association with CD81 tetraspanin, a BCR-coreceptor protein and revealed that IL-27p28 is a transmembrane protein in B cells. Most surprising, we found that i27-Bregs secrete IL-27-containing exosomes (i27-exosomes) and adoptive transfer of i27-exosomes suppressed uveitis by antagonizing Th1/Th17 cells, up-regulating inhibitory-receptors associated with T-cell exhaustion while inducing Treg expansion. Discussion Use of i27-exosomes thus obviates the IL-27 dosing problem, making it possible to determine bioavailable heterodimeric IL-27 needed for therapy. Moreover, as exosomes readily cross the blood-retina-barrier and no adverse effects were observed in mice treated with i27-exosome, results of this study suggest that i27-exosomes might be a promising therapeutic approach for CNS autoimmune diseases.
Collapse
|
15
|
Alves JL, Lemos L, Rodrigues NM, Pereira VB, Barros PAV, Canesso MCC, Guimarães MAF, Cara DC, Miyoshi A, Azevedo VA, Maioli TU, Gomes-Santos AC, Faria AMC. Immunomodulatory effects of different strains of Lactococcus lactis in DSS-induced colitis. Braz J Microbiol 2023; 54:1203-1215. [PMID: 36821043 PMCID: PMC10234881 DOI: 10.1007/s42770-023-00928-0] [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: 12/12/2021] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are gastrointestinal disorders characterized by a breakdown in intestinal homeostasis by inflammatory immune responses to luminal antigens. Novel strategies for ameliorating IBD have been proposed in many studies using animal models. Our group has demonstrated that administration of Lactococcus lactis NCDO 2118 can improve clinical parameters of colitis induced by oral administration of dextran sulphate sodium (DSS). However, it is not clear whether other strains of L. lactis can yield the same effect. The objective of present study was to analyze the effects of three different L. lactis strains (NCDO2118, IL1403 and MG1363) in the development of DSS-induced colitis in C57BL/6 mice. Acute colitis was induced in C57/BL6 mice by the administration of 2% DSS during 7 consecutive days. Body weight loss and shortening of colon length were observed in DSS-treated mice, and none of L. lactis strains had an impact in these clinical signs of colitis. On the other hand, all strains improved the global macroscopical disease index and prevented goblet cells depletion as well as the increase of intestinal permeability. TNF-α production was reduced in gut mucosa of L. lactis DSS-treated mice indicating a modulation of a critical pro-inflammatory response by all strains tested. However, only L. lactis NCDO2118 and MG1363 induced a higher frequency of CD11c+CD11b-CD103+ tolerogenic dendritic cells in lymphoid organs of mice at steady state. We conclude that all tested strains of L. lactis improved the clinical scores and parameters of colitis, which confirm their anti-inflammatory properties in this model of colitis.
Collapse
Affiliation(s)
- Juliana Lima Alves
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| | - Luisa Lemos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Nubia Morais Rodrigues
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Vanessa Bastos Pereira
- Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Ecologia E Evolução, Belo Horizonte, MG, Brazil
| | - Patrícia A Vieira Barros
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Maria Cecília Campos Canesso
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Mauro A F Guimarães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Denise Carmona Cara
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Anderson Miyoshi
- Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Ecologia E Evolução, Belo Horizonte, MG, Brazil
| | - Vasco Ariston Azevedo
- Departamento de Genética, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Ecologia E Evolução, Belo Horizonte, MG, Brazil
| | - Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ana Cristina Gomes-Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ana Maria Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
16
|
Mansourabadi AH, Aghamajidi A, Dorfaki M, Keshavarz F, Shafeghat Z, Moazzeni A, Arab FL, Rajabian A, Roozbehani M, Falak R, Faraji F, Jafari R. B lymphocytes in COVID-19: a tale of harmony and discordance. Arch Virol 2023; 168:148. [PMID: 37119286 PMCID: PMC10147999 DOI: 10.1007/s00705-023-05773-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/29/2023] [Indexed: 05/01/2023]
Abstract
B lymphocytes play a vital role in the human defense against viral infections by producing specific antibodies. They are also critical for the prevention of infectious diseases by vaccination, and their activation influences the efficacy of the vaccination. Since the beginning of coronavirus disease 2019 (COVID-19), which became the main concern of the world health system, many efforts have been made to treat and prevent the disease. However, for the development of successful therapeutics and vaccines, it is necessary to understand the interplay between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, and the immune system. The innate immune system provides primary and nonspecific defense against the virus, but within several days after infection, a virus-specific immune response is provided first by antibody-producing B cells, which are converted after the resolution of disease to memory B cells, which provide long-term immunity. Although a failure in B cell activation or B cell dysfunction can cause a severe form of the disease and also lead to vaccination inefficiency, some individuals with B cell immunodeficiency have shown less production of the cytokine IL-6, resulting in a better disease outcome. In this review, we present the latest findings on the interaction between SARS-CoV-2 and B lymphocytes during COVID-19 infection.
Collapse
Affiliation(s)
- Amir Hossein Mansourabadi
- Department of Immunology, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Dorfaki
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Keshavarz
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Shafeghat
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Moazzeni
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Fahimeh Lavi Arab
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, School of Medicine, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Rajabian
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Faraji
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Institue of Immunology and Infectious diseases, Hazrat-e Rasool General Hospital, Floor 3, Building no. 3, Niyayesh St, Sattar Khan St, 1445613131, Tehran, Iran.
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
- Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St., Ershad Blvd, P.O. Box: 1138, 57147, Urmia, Iran.
| |
Collapse
|
17
|
Daikidou DV, Lioulios G, Sampani E, Xochelli A, Nikolaidou V, Moysidou E, Christodoulou M, Iosifidou A, Iosifidou M, Briza DI, Papagianni A, Fylaktou A, Stangou M. Prospective Analysis of B Lymphocyte Subtypes, before and after Initiation of Dialysis, in Patients with End-Stage Renal Disease. Life (Basel) 2023; 13:life13040860. [PMID: 37109388 PMCID: PMC10146774 DOI: 10.3390/life13040860] [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: 02/12/2023] [Revised: 03/08/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
End-stage renal disease (ESRD) is followed by alterations in adaptive immunity. The aim of this study was to evaluate B lymphocyte subtypes in ESRD patients before and after hemodialysis (HD) or continuous ambulatory peritoneal dialysis (CAPD). PATIENTS AND METHODS CD5, CD27, BAFF, IgM and annexin were evaluated by flow cytometry on CD19+ cells in ESRD patients (n = 40), at time of initiating HD or CAPD (T0) and 6 months later (T6). RESULTS A significant reduction in ESRD-T0 compared to controls was noticed for CD19+, 70.8 (46.5) vs. 171 (249), p < 0.0001, CD19+CD5-, 68.6 (43) vs. 168.9 (106), p < 0.0001, CD19+CD27-, 31.2 (22.1) vs. 59.7 (88.4), p < 0.0001, CD19+CD27+, 42.1 (63.6) vs. 84.3 (78.1), p = 0.002, CD19+BAFF+, 59.7 (37.8) vs. 127.9 (123.7), p < 0.0001 and CD19+IgM+ cells, 48.9 (42.8) vs. 112.5 (81.7) (K/μL), p < 0.0001. The ratio of early/late apoptotic B lymphocytes was reduced (16.8 (10.9) vs. 110 (25.4), p = 0.03). CD19+CD5+ cells were the only cell type with an increased proportion in ESRD-T0 patients (2.7 (3.7) vs. 0.6 (1.1), p < 0.0001). After 6 months on CAPD or HD, CD19+CD27-(%) and early apoptotic lymphocytes were reduced further. The HD patients also showed a significant increase in late apoptotic lymphocytes, from 1.2 (5.7) to 4.2 (7.2) K/mL, p = 0.02. CONCLUSIONS B cells and most of their subtypes were significantly reduced in ESRD-T0 patients compared to controls, the only exception being CD19+CD5+ cells. Apoptotic changes were prominent in ESRD-T0 patients and were exacerbated by HD.
Collapse
Affiliation(s)
- Dimitra-Vasilia Daikidou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece
| | - Georgios Lioulios
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece
| | - Erasmia Sampani
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece
| | - Aliki Xochelli
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Vasiliki Nikolaidou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Eleni Moysidou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece
| | - Michalis Christodoulou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece
| | - Artemis Iosifidou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
| | - Myrto Iosifidou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
| | - Dimitria Ioanna Briza
- School of Informatics, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
| | - Aikaterini Papagianni
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece
| | - Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece
| | - Maria Stangou
- School of Medicine, Aristotle University of Thessaloniki, 45636 Thesaloniki, Greece
- Department of Nephrology, Hippokration Hospital, 54642 Thessaloniki, Greece
| |
Collapse
|
18
|
Barboza BR, Thomaz SMDO, Junior ADC, Espreafico EM, Miyamoto JG, Tashima AK, Camacho MF, Zelanis A, Roque-Barreira MC, da Silva TA. ArtinM Cytotoxicity in B Cells Derived from Non-Hodgkin's Lymphoma Depends on Syk and Src Family Kinases. Int J Mol Sci 2023; 24:ijms24021075. [PMID: 36674590 PMCID: PMC9863955 DOI: 10.3390/ijms24021075] [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: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 01/09/2023] Open
Abstract
Receptors on the immune cell surface have a variety of glycans that may account for the immunomodulation induced by lectins, which have a carbohydrate recognition domain (CRD) that binds to monosaccharides or oligosaccharides in a specific manner. ArtinM, a D-mannose-binding lectin obtained from Artocarpus heterophyllus, has affinity for the N-glycans core. Immunomodulation by ArtinM toward the Th1 phenotype occurs via its interaction with TLR2/CD14 N-glycans on antigen-presenting cells, as well as recognition of CD3γ N-glycans on murine CD4+ and CD8+ T cells. ArtinM exerts a cytotoxic effect on Jurkat human leukemic T-cell line and human myeloid leukemia cell line (NB4). The current study evaluated the effects of ArtinM on murine and human B cells derived from non-Hodgkin’s lymphoma. We found that murine B cells are recognized by ArtinM via the CRD, and the ArtinM stimulus did not augment the proliferation rate or production of IL-2. However, murine B cell incubation with ArtinM augmented the rate of apoptosis, and this cytotoxic effect of ArtinM was also seen in human B cell-lines sourced from non-Hodgkin’s lymphoma Raji cell line. This cytotoxic effect was inhibited by the phosphatase activity of CD45 on Lck, and the protein kinases of the Src family contribute to cell death triggered by ArtinM.
Collapse
Affiliation(s)
- Bruno Rafael Barboza
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Sandra Maria de Oliveira Thomaz
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Airton de Carvalho Junior
- Laboratory of Cell and Molecular Biology of Cancer, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Enilza Maria Espreafico
- Laboratory of Cell and Molecular Biology of Cancer, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Jackson Gabriel Miyamoto
- Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo (EPM/UNIFESP), Sao Paulo 04021-001, SP, Brazil
| | - Alexandre Keiji Tashima
- Department of Biochemistry, Paulista School of Medicine, Federal University of São Paulo (EPM/UNIFESP), Sao Paulo 04021-001, SP, Brazil
| | - Maurício Frota Camacho
- Functional Proteomics Laboratory, Department of Science and Technology, Federal University of São Paulo (ICT-UNIFESP), São José dos Campos 04021-001, SP, Brazil
| | - André Zelanis
- Functional Proteomics Laboratory, Department of Science and Technology, Federal University of São Paulo (ICT-UNIFESP), São José dos Campos 04021-001, SP, Brazil
| | - Maria Cristina Roque-Barreira
- Laboratory of Immunochemistry and Glycobiology, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
| | - Thiago Aparecido da Silva
- Laboratory of Immunotherapy of Invasive Fungal Infections, Department of Cell and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto Medical School, University of São Paulo (FMRP/USP), Ribeirao Preto 14049-900, SP, Brazil
- Correspondence: or ; Tel.: +55-16-3315-3049
| |
Collapse
|
19
|
Dong X, Tu H, Qin S, Bai X, Yang F, Li Z. Insights into the Roles of B Cells in Patients with Sepsis. J Immunol Res 2023; 2023:7408967. [PMID: 37128298 PMCID: PMC10148744 DOI: 10.1155/2023/7408967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/02/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023] Open
Abstract
Sepsis is a life-threatening yet common disease, still posing high mortality worldwide. Sepsis-related deaths primarily occur during immunosuppression; the disease can hamper the numbers and function of B cells, which mediate innate and adaptive immune responses to maintain immune homeostasis. Dysfunction of B cells, along with aggravated immunosuppression, are closely related to poor prognosis. However, B cells in patients with sepsis have garnered little attention. This article focuses on the significance of B-cell subsets, including regulatory B cells, in sepsis and how the counts and function of circulating B cells are affected in patients with sepsis. Finally, potential B-cell-related immunotherapies for sepsis are explored.
Collapse
Affiliation(s)
- Xijie Dong
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Tu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuang Qin
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fan Yang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
20
|
Zimmerman LM. Adaptive Immunity in Reptiles: Conventional Components but Unconventional Strategies. Integr Comp Biol 2022; 62:1572-1583. [PMID: 35482599 DOI: 10.1093/icb/icac022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 01/05/2023] Open
Abstract
Recent studies have established that the innate immune system of reptiles is broad and robust, but the question remains: What role does the reptilian adaptive immune system play? Conventionally, adaptive immunity is described as involving T and B lymphocytes that display variable receptors, is highly specific, improves over the course of the response, and produces a memory response. While reptiles do have B and T lymphocytes that utilize variable receptors, their adaptive response is relatively non-specific, generates a prolonged antibody response, and does not produce a typical memory response. This alternative adaptive strategy may allow reptiles to produce a broad adaptive response that complements a strong innate system. Further studies into reptile adaptive immunity cannot only clarify outstanding questions on the reptilian immune system but can shed light on a number of important immunological concepts, including the evolution of the immune system and adaptive immune responses that take place outside of germinal centers.
Collapse
|
21
|
Field EK, Hartzheim A, Terry J, Dawson G, Haydt N, Neuman-Lee LA. Reptilian Innate Immunology and Ecoimmunology: What Do We Know and Where Are We Going? Integr Comp Biol 2022; 62:1557-1571. [PMID: 35833292 DOI: 10.1093/icb/icac116] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 01/05/2023] Open
Abstract
Reptiles, the only ectothermic amniotes, employ a wide variety of physiological adaptations to adjust to their environments but remain vastly understudied in the field of immunology and ecoimmunology in comparison to other vertebrate taxa. To address this knowledge gap, we assessed the current state of research on reptilian innate immunology by conducting an extensive literature search of peer-reviewed articles published across the four orders of Reptilia (Crocodilia, Testudines, Squamata, and Rhynchocephalia). Using our compiled dataset, we investigated common techniques, characterization of immune components, differences in findings and type of research among the four orders, and immune responses to ecological and life-history variables. We found that there are differences in the types of questions asked and approaches used for each of these reptilian orders. The different conceptual frameworks applied to each group has led to a lack of unified understanding of reptilian immunological strategies, which, in turn, have resulted in large conceptual gaps in the field of ecoimmunology as a whole. To apply ecoimmunological concepts and techniques most effectively to reptiles, we must combine traditional immunological studies with ecoimmunological studies to continue to identify, characterize, and describe the reptilian immune components and responses. This review highlights the advances and gaps that remain to help identify targeted and cohesive approaches for future research in reptilian ecoimmunological studies.
Collapse
Affiliation(s)
- Emily K Field
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Alyssa Hartzheim
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Jennifer Terry
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Grant Dawson
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Natalie Haydt
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| | - Lorin A Neuman-Lee
- Department of Biological Sciences, Arkansas State University, Jonesboro, AR 72401, USA
| |
Collapse
|
22
|
Ma C, Liu H, Yang S, Li H, Liao X, Kang Y. The emerging roles and therapeutic potential of B cells in sepsis. Front Pharmacol 2022; 13:1034667. [PMID: 36425582 PMCID: PMC9679374 DOI: 10.3389/fphar.2022.1034667] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/26/2022] [Indexed: 01/03/2024] Open
Abstract
Sepsis is a life-threatening syndrome caused by anomalous host response to infection. The pathogenesis of sepsis is complex, and immune dysfunction is the central link in its occurrence and development. The sepsis immune response is not a local and transient process but a complex and continuous process involving all major cell types of innate and adaptive immunity. B cells are traditionally studied for their ability to produce antibodies in the context of mediating humoral immunity. However, over the past few years, B cells have been increasingly recognized as key modulators of adaptive and innate immunity, and they can participate in immune responses by presenting antigens, producing cytokines, and modulating other immune cells. Recently, increasing evidence links B-cell dysfunction to mechanisms of immune derangement in sepsis, which has drawn attention to the powerful properties of this unique immune cell type in sepsis. Here, we reviewed the dynamic alterations of B cells and their novel roles in animal models and patients with sepsis, and provided new perspectives for therapeutic strategies targeting B cells in sepsis.
Collapse
Affiliation(s)
- Chengyong Ma
- Center of Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hanrui Liu
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shuo Yang
- Center of Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hong Li
- Center of Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xuelian Liao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Guillamat-Prats R, Hering D, Derle A, Rami M, Härdtner C, Santovito D, Rinne P, Bindila L, Hristov M, Pagano S, Vuilleumier N, Schmid S, Janjic A, Enard W, Weber C, Maegdefessel L, Faussner A, Hilgendorf I, Steffens S. GPR55 in B cells limits atherosclerosis development and regulates plasma cell maturation. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1056-1071. [PMID: 36523570 PMCID: PMC7613934 DOI: 10.1038/s44161-022-00155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 09/27/2022] [Indexed: 06/17/2023]
Abstract
Dissecting the pathways regulating the adaptive immune response in atherosclerosis is of particular therapeutic interest. Here we report that the lipid G-protein coupled receptor GPR55 is highly expressed by splenic plasma cells (PC), upregulated in mouse spleens during atherogenesis and human unstable or ruptured compared to stable plaques. Gpr55-deficient mice developed larger atherosclerotic plaques with increased necrotic core size compared to their corresponding controls. Lack of GPR55 hyperactivated B cells, disturbed PC maturation and resulted in immunoglobulin (Ig)G overproduction. B cell-specific Gpr55 depletion or adoptive transfer of Gpr55-deficient B cells was sufficient to promote plaque development and elevated IgG titers. In vitro, the endogenous GPR55 ligand lysophsophatidylinositol (LPI) enhanced PC proliferation, whereas GPR55 antagonism blocked PC maturation and increased their mitochondrial content. Collectively, these discoveries provide previously undefined evidence for GPR55 in B cells as a key modulator of the adaptive immune response in atherosclerosis.
Collapse
Affiliation(s)
- Raquel Guillamat-Prats
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Daniel Hering
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Abhishek Derle
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Martina Rami
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Carmen Härdtner
- Department of Cardiology and Angiology I, Heart Center and Faculty of Medicine, University of Freiburg. Freiburg, Germany
| | - Donato Santovito
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Institute for Genetic and Biomedical Research (IRGB), Unit of Milan, National Research Council, Milan, Italy
| | - Petteri Rinne
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura Bindila
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michael Hristov
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Sabrina Pagano
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals and Faculty of Medicine
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals and Faculty of Medicine
| | - Sofie Schmid
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich (TUM), Munich, Germany
| | - Aleksandar Janjic
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Martinsried, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians University, Martinsried, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Lars Maegdefessel
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
- Department of Vascular and Endovascular Surgery, Klinikum rechts der Isar - Technical University Munich (TUM), Munich, Germany
| | - Alexander Faussner
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, Heart Center and Faculty of Medicine, University of Freiburg. Freiburg, Germany
- Institute for Experimental Cardiovascular Medicine, Heart Center and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance (MHA), Munich, Germany
| |
Collapse
|
24
|
Català C, Velasco-de Andrés M, Leyton-Pereira A, Casadó-Llombart S, Sáez Moya M, Gutiérrez-Cózar R, García-Luna J, Consuegra-Fernández M, Isamat M, Aranda F, Martínez-Florensa M, Engel P, Mourglia-Ettlin G, Lozano F. CD6 deficiency impairs early immune response to bacterial sepsis. iScience 2022; 25:105078. [PMID: 36157587 PMCID: PMC9490029 DOI: 10.1016/j.isci.2022.105078] [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: 03/22/2022] [Revised: 07/15/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
CD6 is a lymphocyte-specific scavenger receptor expressed on adaptive (T) and innate (B1a, NK) immune cells, which is involved in both fine-tuning of lymphocyte activation/differentiation and recognition of bacterial-associated molecular patterns (i.e., lipopolysaccharide). However, evidence on CD6’s role in the physiological response to bacterial infection was missing. Our results show that induction of monobacterial and polymicrobial sepsis in Cd6−/− mice results in lower survival rates and increased bacterial loads and pro-inflammatory cytokine levels. Steady state analyses of Cd6−/− mice show decreased levels of natural polyreactive antibodies, concomitant with decreased cell counts of spleen B1a and marginal zone B cells. Adoptive transfer of wild-type B cells and mouse serum, as well as a polyreactive monoclonal antibody improve Cd6−/− mouse survival rates post-sepsis. These findings support a nonredundant role for CD6 in the early response against bacterial infection, through homeostatic expansion and functionality of innate-related immune cells. CD6 is a nonredundant receptor in early immune response to sepsis Cd6−/− mice show higher susceptibility to bacterial sepsis Cd6−/− mice show lower B1a and MZB cell and natural polyreactive antibody levels B cell and serum transfer restore susceptibility of Cd6−/− mice to bacterial sepsis
Collapse
Affiliation(s)
- Cristina Català
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain
| | - María Velasco-de Andrés
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain
| | - Alejandra Leyton-Pereira
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain
| | - Sergi Casadó-Llombart
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain
| | - Manuel Sáez Moya
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Rebeca Gutiérrez-Cózar
- Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Joaquín García-Luna
- Área Inmunología, Facultad de Química/Facultad de Ciencias, DEPBIO/IQB, Universidad de la República, 11800 Montevideo, Uruguay
| | - Marta Consuegra-Fernández
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain
| | - Marcos Isamat
- Sepsia Therapeutics S.L., 08908 L'Hospitalet de Llobregat, Spain
| | - Fernando Aranda
- Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
| | - Mario Martínez-Florensa
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain
| | - Pablo Engel
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain.,Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | - Gustavo Mourglia-Ettlin
- Área Inmunología, Facultad de Química/Facultad de Ciencias, DEPBIO/IQB, Universidad de la República, 11800 Montevideo, Uruguay
| | - Francisco Lozano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Rosselló 149-153, 08036 Barcelona, Spain.,Departament de Biomedicina, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain.,Servei d'Immunologia, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
25
|
Smirnova NF, Riemondy K, Bueno M, Collins S, Suresh P, Wang X, Patel KN, Cool C, Königshoff M, Sharma NS, Eickelberg O. Single-cell transcriptome mapping identifies a local, innate B cell population driving chronic rejection after lung transplantation. JCI Insight 2022; 7:156648. [PMID: 36134664 PMCID: PMC9675462 DOI: 10.1172/jci.insight.156648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is the main reason for poor outcomes after lung transplantation (LTx). We and others have recently identified B cells as major contributors to BOS after LTx. The extent of B cell heterogeneity and the relative contributions of B cell subpopulations to BOS, however, remain unclear. Here, we provide a comprehensive analysis of cell population changes and their gene expression patterns during chronic rejection after orthotopic LTx in mice. Of 11 major cell types, Mzb1-expressing plasma cells (PCs) were the most prominently increased population in BOS lungs. These findings were validated in 2 different cohorts of human BOS after LTx. A Bhlhe41, Cxcr3, and Itgb1 triple-positive B cell subset, also expressing classical markers of the innate-like B-1 B cell population, served as the progenitor pool for Mzb1+ PCs. This subset accounted for the increase in IgG2c production within BOS lung grafts. A genetic lack of Igs decreased BOS severity after LTx. In summary, we provide a detailed analysis of cell population changes during BOS. IgG+ PCs and their progenitors — an innate B cell subpopulation — are the major source of local Ab production and a significant contributor to BOS after LTx.
Collapse
Affiliation(s)
- Natalia F Smirnova
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Institut des Maladies Métaboliques et Cardiovasculaires (I2MC) - INSERM U1297, University of Toulouse III, Toulouse, France
| | - Kent Riemondy
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Marta Bueno
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Susan Collins
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Pavan Suresh
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Xingan Wang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kapil N Patel
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida/Tampa General Hospital, Tampa, Florida, USA
| | - Carlyne Cool
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melanie Königshoff
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nirmal S Sharma
- Center for Advanced Lung Disease and Lung Transplantation, University of South Florida/Tampa General Hospital, Tampa, Florida, USA.,Division of Pulmonary & Critical Care, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
26
|
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
|
27
|
Valeff NJ, Ventimiglia MS, Dibo M, Markert UR, Jensen F. Splenic B1 B Cells Acquire a Proliferative and Anti-Inflamatory Profile During Pregnancy in Mice. Front Immunol 2022; 13:873493. [PMID: 35572585 PMCID: PMC9095819 DOI: 10.3389/fimmu.2022.873493] [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: 02/10/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
B cells are a heterogeneous cell population with differential ontogeny, anatomical location, and functions. B1 B cells are a distinct subpopulation characterized by their unique capacity of self-renewal, the production of large quantities of IL-10, and the ability to secrete protective, anti-inflammatory natural antibodies (NAbs), presumably upon down-regulation of CD1d expression. Although natural antibodies are thought to be protective, due to their polyreactivity, their participation in certain autoimmune diseases has been suggested. In the context of pregnancy, the role of B1 B cells has been discussed controversially. While in human pregnancies B1 B cells and natural/polyreactive antibodies they produce are involved in the development of preeclampsia, in mice they promote healthy gestation and fetal protection. In this work, we aimed to functionally characterize the splenic B1 B cell population during pregnancy in mice. Functional enrichment analysis using only up-regulated transcripts from a transcriptomic profile performed on total splenic B cells from pregnant compared to non-pregnant mice showed augmented cell cycle and DNA replication pathways. Proliferation studies by flow cytometry showed augmented Ki-67 proliferation marker expression and percentages of B1 B cells. Furthermore, B1 B cells produced higher levels of IL-10 and lower levels of TNF-α leading to an increased IL-10/TNF-α ratio and showing an immunoregulatory phenotype. Finally, we observed lower expression of CD1d on B1 B cells, suggesting a higher capacity to produce NAbs in the context of pregnancy. In summary, our results showed not only an expanded and proliferative splenic B1 B cell population during pregnancy but also the acquisition of immunomodulatory capacities suggesting its critical role in the intricate process of pregnancy tolerance.
Collapse
Affiliation(s)
- Natalin J Valeff
- Laboratorio de Inmunología de la Reproducción, CEFYBO-UBA-CONICET, Buenos Aires, Argentina
| | - María S Ventimiglia
- Laboratorio de Inmunología de la Reproducción, CEFYBO-UBA-CONICET, Buenos Aires, Argentina
| | - Marcos Dibo
- Laboratorio de Inmunología de la Reproducción, CEFYBO-UBA-CONICET, Buenos Aires, Argentina
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Jena, Germany
| | - Federico Jensen
- Laboratorio de Inmunología de la Reproducción, CEFYBO-UBA-CONICET, Buenos Aires, Argentina.,Centro Integrativo de Biología Y Química Aplicada, Universidad Bernardo O'Higgins, Santiago, Chile
| |
Collapse
|
28
|
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: 11] [Impact Index Per Article: 3.7] [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
|
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: 6] [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
|
Ordoñez R, Solano D, Granizo G. T and B Cells Immune Response and the importance of vaccines Against SARS-CoV-2. BIONATURA 2022. [DOI: 10.21931/rb/2022.07.01.36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 has become a global pandemic because it is a severe respiratory syndrome that attacks many people worldwide and can lead to death depending on the severity. In recent years, the study of the acquired immune response (T cells) and innate (B cells) has increased to better treat the disease from the quantitative cell count. A picture has begun to emerge revealing that CD4+ T cells, CD8+ T cells, and neutralizing antibodies contribute to the control of SARS-CoV-2 in COVID-19 cases. This work studies the three fundamental components of the adaptive immune system: B cells (the source of antibodies), CD4+T cells, and CD8+T cells and their function against SARS-CoV2. The importance of vaccines and the different types of existing vaccines are discussed. Implications of covid-19 variants on Immunity and vaccine types are also analyzed to understand how the action of the immune system will help treat the disease.
Collapse
Affiliation(s)
- Ronny Ordoñez
- School of Biological Science and Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí 100119, Ecuador
| | - Dulexy Solano
- School of Biological Science and Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí 100119, Ecuador
| | - Gustavo Granizo
- School of Biological Science and Engineering, Yachay Tech University, Hacienda San José s/n, San Miguel de Urcuquí 100119, Ecuador
| |
Collapse
|
31
|
Early Reconstitution of Antibody Secreting Cells after Allogeneic Stem Cell Transplantation. J Clin Med 2022; 11:jcm11010270. [PMID: 35012014 PMCID: PMC8745805 DOI: 10.3390/jcm11010270] [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: 11/25/2021] [Revised: 12/26/2021] [Accepted: 01/03/2022] [Indexed: 11/24/2022] Open
Abstract
Immune cell reconstitution after stem cell transplantation is allocated over several stages. Whereas cells mediating innate immunity recover rapidly, adaptive immune cells, including T and B cells, recover slowly over several months. In this study we investigated kinetics and reconstitution of de novo B cell formation in patients receiving CD3 and CD19 depleted haploidentical stem cell transplantation with additional in vivo T cell depletion with monoclonal anti-CD3 antibody. This model enables a detailed in vivo evaluation of hierarchy and attribution of defined lymphocyte populations without skewing by mTOR- or NFAT-inhibitors. As expected CD3+ T cells and their subsets had delayed reconstitution (<100 cells/μL at day +90). Well defined CD19+ B lymphocytes of naïve and memory phenotype were detected at day +60. Remarkably, we observed a very early reconstitution of antibody-secreting cells (ASC) at day +14. These ASC carried the HLA-haplotype of the donor and secreted the isotypes IgM and IgA more prevalent than IgG. They correlated with a population of CD19− CD27− CD38low/+ CD138− cells. Of note, reconstitution of this ASC occurred without detectable circulating T cells and before increase of BAFF or other B cell stimulating factors. In summary, we describe a rapid reconstitution of peripheral blood ASC after CD3 and CD19 depleted haploidentical stem cell transplantation, far preceding detection of naïve and memory type B cells. Incidence before T cell reconstitution and spontaneous secretion of immunoglobulins allocate these early ASC to innate immunity, eventually maintaining natural antibody levels.
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: 0.8] [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
|
Hou Y, Li Y, Liu B, Wan H, Liu C, Xia W. nnResearch progress on B cells and thoracic aortic aneurysm/dissection. Ann Vasc Surg 2021; 82:377-382. [PMID: 34933111 DOI: 10.1016/j.avsg.2021.11.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/23/2021] [Accepted: 11/28/2021] [Indexed: 02/02/2023]
Abstract
Thoracic aortic aneurysm/dissection (TAAD) is a rare cardiovascular disease characterized by acute onset, rapid progression and high morbidity and mortality. One of the crucial factors leading to TAAD is the inflammatory response, which is regulated by many immune cell subgroups, including B cells. Compared with normal aortic tissue, the number of B cells in the aortic tissue of TAAD patients is significantly higher. Activated B cells participate in the vascular immune inflammatory response by producing antibodies and inflammatory factors and activating the complement system. These effects can lead to collagen degradation and aortic wall remodeling, both of which are the main pathologic characteristics of TAAD. Therefore, B cells play a key role in the occurrence and development of TAAD. B cells can be divided into B1 cells, B2 cells and regulatory B cells, which have different mechanisms of action in TAAD. This article will review the role of B cells in TAAD from the perspective of three different subtypes of B cells.
Collapse
Affiliation(s)
- Yue Hou
- Clinical laboratory diagnostics, Beihua University, China
| | - Yan Li
- Clinical laboratory diagnostics, Beihua University, China
| | - Bingqing Liu
- Clinical laboratory diagnostics, Beihua University, China
| | - Hong Wan
- Clinical laboratory diagnostics, Beihua University, China
| | - Chang Liu
- Clinical laboratory diagnostics, Beihua University, China.
| | - Wei Xia
- Clinical laboratory diagnostics, Beihua University, China.
| |
Collapse
|
34
|
Ramírez-de-Arellano A, Gutiérrez-Franco J, Sierra-Diaz E, Pereira-Suárez AL. The role of estradiol in the immune response against COVID-19. Hormones (Athens) 2021; 20:657-667. [PMID: 34142358 PMCID: PMC8210971 DOI: 10.1007/s42000-021-00300-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the pathogen agent causing coronavirus disease (COVID)-19, which was declared a global pandemic in 2020. The spike protein of this virus and the angiotensin-converter enzyme (ACE)-2 in host cells in humans play a vital role in infection and in COVID-19 pathogenesis. Estradiol is known to modulate the actions of immune cells, and, therefore, the antiviral mechanisms of these cells could also be modified by this hormone stimulus. Even though estradiol is not considered a protective factor, evidence shows that women with high levels of this hormone have a lower risk of developing severe symptoms and an even a lower incidence of death. Understanding the mechanism of action of estradiol with regard to viral infections and COVID-19 is essential for the improvement of therapeutic strategies. This review aims to describe the effects that estradiol exerts on immune cells during viral infections and COVID-19.
Collapse
Affiliation(s)
- Adrián Ramírez-de-Arellano
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Jalisco, 44 340, México
| | - Jorge Gutiérrez-Franco
- Unidad Académica de Ciencias Químico Biológicas Y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, 63 000, México
| | - Erick Sierra-Diaz
- Departamento de Salud Pública, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, División de Epidemiología, Unidad Medica de Alta Especialidad, Hospital de Especialidades, Centro Médico Nacional de Occidente, Guadalajara, Jalisco, 44340, México
| | - Ana Laura Pereira-Suárez
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Guadalajara, Jalisco, 44 340, México.
- Departamento de Microbiología Y Patología, Centro Universitario de Ciencias de La Salud, Universidad de Guadalajara, Sierra Mojada # 950, Colonia Independencia, CP, 44340, Guadalajara, Jalisco, México.
| |
Collapse
|
35
|
IL-27-producing B-1a cells suppress neuroinflammation and CNS autoimmune diseases. Proc Natl Acad Sci U S A 2021; 118:2109548118. [PMID: 34782464 DOI: 10.1073/pnas.2109548118] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2021] [Indexed: 01/06/2023] Open
Abstract
Regulatory B cells (Breg cells) that secrete IL-10 or IL-35 (i35-Breg) play key roles in regulating immunity in tumor microenvironment or during autoimmune and infectious diseases. Thus, loss of Breg function is implicated in development of autoimmune diseases while aberrant elevation of Breg prevents sterilizing immunity, exacerbates infectious diseases, and promotes cancer metastasis. Breg cells identified thus far are largely antigen-specific and derive mainly from B2-lymphocyte lineage. Here, we describe an innate-like IL-27-producing natural regulatory B-1a cell (i27-Breg) in peritoneal cavity and human umbilical cord blood. i27-Bregs accumulate in CNS and lymphoid tissues during neuroinflammation and confers protection against CNS autoimmune disease. i27-Breg immunotherapy ameliorated encephalomyelitis and uveitis through up-regulation of inhibitory receptors (Lag3, PD-1), suppression of Th17/Th1 responses, and propagating inhibitory signals that convert conventional B cells to regulatory lymphocytes that secrete IL-10 and/or IL-35 in eye, brain, or spinal cord. Furthermore, i27-Breg proliferates in vivo and sustains IL-27 secretion in CNS and lymphoid tissues, a therapeutic advantage over administering biologics (IL-10, IL-35) that are rapidly cleared in vivo. Mutant mice lacking irf4 in B cells exhibit exaggerated increase of i27-Bregs with few i35-Bregs, while mice with loss of irf8 in B cells have abundance of i35-Bregs but defective in generating i27-Bregs, identifying IRF8/BATF and IRF4/BATF axis in skewing B cell differentiation toward i27-Breg and i35-Breg developmental programs, respectively. Consistent with its developmental origin, disease suppression by innate i27-Bregs is neither antigen-specific nor disease-specific, suggesting that i27-Breg would be effective immunotherapy for a wide spectrum of autoimmune diseases.
Collapse
|
36
|
Badger R, Park K, Pietrofesa RA, Christofidou-Solomidou M, Serve KM. Late Inflammation Induced by Asbestiform Fibers in Mice Is Ameliorated by a Small Molecule Synthetic Lignan. Int J Mol Sci 2021; 22:ijms222010982. [PMID: 34681644 PMCID: PMC8537122 DOI: 10.3390/ijms222010982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Exposure to Libby amphibole (LA) asbestos-like fibers is associated with increased risk of asbestosis, mesothelioma, pulmonary disease, and systemic autoimmune disease. LGM2605 is a small molecule antioxidant and free radical scavenger, with anti-inflammatory effects in various disease models. The current study aimed to determine whether the protective effects of LGM2605 persist during the late inflammatory phase post-LA exposure. Male and female C57BL/6 mice were administered daily LGM2605 (100 mg/kg) via gel cups for 3 days before and 14 days after a 200 µg LA given via intraperitoneal (i.p.) injection. Control mice were given unsupplemented gel cups and an equivalent dose of i.p. saline. On day 14 post-LA treatment, peritoneal lavage was assessed for immune cell influx, cytokine concentrations, oxidative stress biomarkers, and immunoglobulins. During the late inflammatory phase post-LA exposure, we noted an alteration in trafficking of both innate and adaptive immune cells, increased pro-inflammatory cytokine concentrations, induction of immunoglobulin isotype switching, and increased oxidized guanine species. LGM2605 countered these changes similarly among male and female mice, ameliorating late inflammation and altering immune responses in late post-LA exposure. These data support possible efficacy of LGM2605 in the prolonged treatment of LA-associated disease and other inflammatory conditions.
Collapse
Affiliation(s)
- Reagan Badger
- Department of Biological Sciences, Idaho State University, Pocatello, ID 83209, USA;
| | - Kyewon Park
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (K.P.); (R.A.P.); (M.C.-S.)
| | - Ralph A. Pietrofesa
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (K.P.); (R.A.P.); (M.C.-S.)
| | - Melpo Christofidou-Solomidou
- Division of Pulmonary, Allergy, and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (K.P.); (R.A.P.); (M.C.-S.)
| | - Kinta M. Serve
- Department of Biological Sciences, Idaho State University, Pocatello, ID 83209, USA;
- Correspondence:
| |
Collapse
|
37
|
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
|
38
|
Azevedo Portilho N, Scarfò R, Bertesago E, Ismailoglu I, Kyba M, Kobayashi M, Ditadi A, Yoshimoto M. B1 lymphocytes develop independently of Notch signaling during mouse embryonic development. Development 2021; 148:271231. [PMID: 34370006 PMCID: PMC8380455 DOI: 10.1242/dev.199373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 07/08/2021] [Indexed: 11/20/2022]
Abstract
B1 lymphocytes are a small but unique component of the innate immune-like cells. However, their ontogenic origin is still a matter of debate. Although it is widely accepted that B1 cells originate early in fetal life, whether or not they arise from hematopoietic stem cells (HSCs) is still unclear. In order to shed light on the B1 cell origin, we set out to determine whether their lineage specification is dependent on Notch signaling, which is essential for the HSC generation and, therefore, all derivatives lineages. Using mouse embryonic stem cells (mESCs) to recapitulate murine embryonic development, we have studied the requirement for Notch signaling during the earliest B-cell lymphopoiesis and found that Rbpj-deficient mESCs are able to generate B1 cells. Their Notch independence was confirmed in ex vivo experiments using Rbpj-deficient embryos. In addition, we found that upregulation of Notch signaling induced the emergence of B2 lymphoid cells. Taken together, these findings indicate that control of Notch signaling dose is crucial for different B-cell lineage specification from endothelial cells and provides pivotal information for their in vitro generation from PSCs for therapeutic applications. This article has an associated ‘The people behind the papers’ interview. Summary: During development, the first B lymphocytes are produced in the absence of Notch signaling and, thus, independently from HSCs.
Collapse
Affiliation(s)
- Nathalia Azevedo Portilho
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Rebecca Scarfò
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Elisa Bertesago
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Ismail Ismailoglu
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA.,Genetics and Development Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael Kyba
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michihiro Kobayashi
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Andrea Ditadi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Momoko Yoshimoto
- Center for Stem Cell Research and Regenerative Medicine, Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
39
|
Royster W, Jin H, Wang P, Aziz M. Extracellular CIRP decreases Siglec-G expression on B-1a cells skewing them towards a pro-inflammatory phenotype in sepsis. Mol Med 2021; 27:55. [PMID: 34058975 PMCID: PMC8165807 DOI: 10.1186/s10020-021-00318-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Sepsis is a life-threatening disease syndrome caused by a dysregulated host response to infection and injury. Extracellular cold-inducible RNA-binding protein (eCIRP) acts as a damage-associated molecular pattern. Peritoneal cavity (PerC) B-1a cells attenuate inflammation and tissue injury by spontaneous releasing natural IgM and IL-10. Sialic acid-binding immunoglobulin-type lectin-G (Siglec-G) is a CD33-related receptor highly expressed in B-1a cells to serve critical immunoregulatory functions. In sepsis, B-1a cell numbers in PerC are decreased. We hypothesized that eCIRP causes the reduction of PerC B-1a cells and alters their function during sepsis. METHODS Sepsis was induced in WT and CIRP-/- mice by cecal ligation and puncture (CLP). PerC washout cells were collected and B-1a cells and Siglec-G were assessed by flow cytometry. Mice were i.p. injected with recombinant murine (rm) CIRP and after 20 h, Siglec-G expression in PerC B-1a cells were assessed. PerC B-1a cells were treated with rmCIRP for 4 h and Siglec-G expression was assessed. PerC B-1a cells were pre-treated with anti-Siglec-G Ab and then after stimulated with rmCIRP for 24 h, IL-6 levels in the culture supernatants were assessed. RESULTS eCIRP levels in the PerC were elevated in septic mice. In WT mice, the frequencies and numbers of total and Siglec-G+ B-1a cells in the PerC were significantly decreased in the CLP group compared to sham group, whereas in CIRP-/- mice, their frequencies and numbers in sepsis were significantly rescued compared to WT septic mice. Mice injected with rmCIRP showed decreased frequencies and numbers of total and Siglec-G+ PerC B-1a cells compared to PBS-injected mice. In vitro treatment of PerC B-1a cells with rmCIRP demonstrated significant reduction in Siglec-G mRNA and protein compared to PBS group. PerC B-1a cells treated with anti-Siglec-G Ab had significantly higher production of IL-6 in response to rmCIRP compared to IgG control. Anti-Siglec-G Ab treated B-1a cells co-cultured with macrophages produced significantly higher levels of IL-6, and TNF-α, and lower levels of IL-10 compared to IgG-treated B-1a cells and macrophage co-cultures stimulated with rmCIRP. CONCLUSION eCIRP reduces PerC B-1a cell pool and skews them to a pro-inflammatory phenotype by downregulating Siglec-G expression. Targeting eCIRP will retain Siglec-G expressing B-1a cells in the PerC and preserve their anti-inflammatory function in sepsis.
Collapse
Affiliation(s)
- William Royster
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, 11030, USA
- Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Hui Jin
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, 11030, USA.
- Department of Surgery, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, 350 Community Dr, Manhasset, NY, 11030, USA.
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine At Hofstra/Northwell, Manhasset, NY, 11030, USA.
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, 11030, USA.
| |
Collapse
|
40
|
Liu Y, Hu JN, Luo N, Zhao J, Liu SC, Ma T, Yao YM. The Essential Involvement of the Omentum in the Peritoneal Defensive Mechanisms During Intra-Abdominal Sepsis. Front Immunol 2021; 12:631609. [PMID: 33815381 PMCID: PMC8012523 DOI: 10.3389/fimmu.2021.631609] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/03/2021] [Indexed: 11/16/2022] Open
Abstract
Although the abilities of the omentum to alleviate inflammation and prevent infection have been revealed over the past decades, the underlying mechanisms remain largely unelucidated. Here, we demonstrated that the mortality of mice exposed to cecal ligation and puncture (CLP) and omentectomy was remarkably increased compared to those treated with CLP alone. Moreover, the efficacy of the omentum was associated with an impairment in intraperitoneal bacterial clearance together with an increase in the expression of proinflammatory cytokines. Besides, in response to peritoneal infections, the size and quantity of the omental milky spots (MSs) were increased tremendously and they also support innate-like B1 cell responses and local IgM production in the peritoneal cavity. Furthermore, not only the migration but also the functional activities of neutrophils were diminished in the absence of the omentum. These data collectively show that the omentum contributes more to peritoneal immune responses during septic peritonitis than has heretofore been recognized. Thus, harnessing the function of MS-containing omentum to increase its protective effectiveness may exert important biological and therapeutic implications for the control of intra-abdominal infections.
Collapse
Affiliation(s)
- Ying Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jian-Nan Hu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ning Luo
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jie Zhao
- Department of Intensive Care Unit, Tianjin Medical University General Hospital, Tianjin, China
| | - Shu-Chang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Ma
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Yong-Ming Yao
- Department of Microbiology and Immunology, Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
41
|
Bednarczyk M, Medina-Montano C, Fittler FJ, Stege H, Roskamp M, Kuske M, Langer C, Vahldieck M, Montermann E, Tubbe I, Röhrig N, Dzionek A, Grabbe S, Bros M. Complement-Opsonized Nano-Carriers Are Bound by Dendritic Cells (DC) via Complement Receptor (CR)3, and by B Cell Subpopulations via CR-1/2, and Affect the Activation of DC and B-1 Cells. Int J Mol Sci 2021; 22:2869. [PMID: 33799879 PMCID: PMC8001596 DOI: 10.3390/ijms22062869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The development of nanocarriers (NC) for biomedical applications has gained large interest due to their potential to co-deliver drugs in a cell-type-targeting manner. However, depending on their surface characteristics, NC accumulate serum factors, termed protein corona, which may affect their cellular binding. We have previously shown that NC coated with carbohydrates to enable biocompatibility triggered the lectin-dependent complement pathway, resulting in enhanced binding to B cells via complement receptor (CR)1/2. Here we show that such NC also engaged all types of splenic leukocytes known to express CR3 at a high rate when NC were pre-incubated with native mouse serum resulting in complement opsonization. By focusing on dendritic cells (DC) as an important antigen-presenting cell type, we show that CR3 was essential for binding/uptake of complement-opsonized NC, whereas CR4, which in mouse is specifically expressed by DC, played no role. Further, a minor B cell subpopulation (B-1), which is important for first-line pathogen responses, and co-expressed CR1/2 and CR3, in general, engaged NC to a much higher extent than normal B cells. Here, we identified CR-1/2 as necessary for binding of complement-opsonized NC, whereas CR3 was dispensable. Interestingly, the binding of complement-opsonized NC to both DC and B-1 cells affected the expression of activation markers. Our findings may have important implications for the design of nano-vaccines against infectious diseases, which codeliver pathogen-specific protein antigen and adjuvant, aimed to induce a broad adaptive cellular and humoral immune response by inducing cytotoxic T lymphocytes that kill infected cells and pathogen-neutralizing antibodies, respectively. Decoration of nano-vaccines either with carbohydrates to trigger complement activation in vivo or with active complement may result in concomitant targeting of DC and B cells and thereby may strongly enhance the extent of dual cellular/humoral immune responses.
Collapse
Affiliation(s)
- Monika Bednarczyk
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Frederic Julien Fittler
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Meike Roskamp
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Michael Kuske
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Christian Langer
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Marco Vahldieck
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Evelyn Montermann
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Ingrid Tubbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Andrzej Dzionek
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| |
Collapse
|
42
|
Royster W, Wang P, Aziz M. The Role of Siglec-G on Immune Cells in Sepsis. Front Immunol 2021; 12:621627. [PMID: 33708213 PMCID: PMC7940683 DOI: 10.3389/fimmu.2021.621627] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/13/2021] [Indexed: 12/30/2022] Open
Abstract
Sepsis is a life-threatening clinical syndrome that results from an overwhelming immune response to infection. During sepsis, immune cells are activated by sensing pathogen-associated molecular patterns and damage-associated molecular patterns (DAMPs) through pattern recognizing receptors (PRRs). Regulation of the immune response is essential to preventing or managing sepsis. Sialic acid-binding immunoglobulin-type lectin-G (Siglec-G), a CD33 group of Siglec expressed in B-1a cells and other hematopoietic cells, plays an important immunoregulatory role. B-1a cells, a subtype of B lymphocytes, spontaneously produce natural IgM which confers protection against infection. B-1a cells also produce IL-10, GM-CSF, and IL-35 to control inflammation. Sialic acids are present on cell membranes, receptors, and glycoproteins. Siglec-G binds to the sialic acid residues on the B cell receptor (BCR) and controls BCR-mediated signal transduction, thereby maintaining homeostasis of Ca++ influx and NFATc1 expression. Siglec-G inhibits NF-κB activation in B-1a cells and regulates B-1a cell proliferation. In myeloid cells, Siglec-G inhibits DAMP-mediated inflammation by forming a ternary complex with DAMP and CD24. Thus, preserving Siglec-G’s function could be a novel therapeutic approach in sepsis. Here, we review the immunoregulatory functions of Siglec-G in B-1a cells and myeloid cells in sepsis. A clear understanding of Siglec-G is important to developing novel therapeutics in treating sepsis.
Collapse
Affiliation(s)
- William Royster
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
| |
Collapse
|
43
|
P2Y 2 receptor antagonism resolves sialadenitis and improves salivary flow in a Sjögren's syndrome mouse model. Arch Oral Biol 2021; 124:105067. [PMID: 33561807 DOI: 10.1016/j.archoralbio.2021.105067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Sjögren's syndrome (SS) is a chronic autoimmune exocrinopathy characterized by lymphocytic infiltration of the salivary and lacrimal glands and decreased saliva and tear production. Previous studies indicate that the G protein-coupled P2Y2 nucleotide receptor (P2Y2R) is upregulated in numerous models of salivary gland inflammation (i.e., sialadenitis), where it has been implicated as a key mediator of chronic inflammation. Here, we evaluate both systemic and localized P2Y2R antagonism as a means to resolve sialadenitis in the NOD.H-2h4,IFNγ-/-,CD28-/- (NOD.H-2h4 DKO) mouse model of SS. DESIGN Female 4.5 month old NOD.H-2h4 DKO mice received daily intraperitoneal injections for 10 days of the selective P2Y2R antagonist, AR-C118925, or vehicle-only control. Single-dose localized intraglandular antagonist delivery into the Wharton's duct was also evaluated. Carbachol-induced saliva was measured and then submandibular glands (SMGs) were isolated and either fixed and paraffin-embedded for H&E staining, homogenized for RNA isolation or dissociated for flow cytometry analysis. RESULTS Intraperitoneal injection, but not localized intraglandular administration, of AR-C118925 significantly enhanced carbachol-induced salivation and reduced lymphocytic foci and immune cell markers in SMGs of 5 month old NOD.H-2h4 DKO mice, compared to vehicle-injected control mice. We found that B cells represent the primary immune cell population in inflamed SMGs of NOD.H-2h4 DKO mice that express elevated levels of P2Y2R compared to C57BL/6 control mice. We further demonstrate a role for P2Y2Rs in mediating B cell migration and the release of IgM. CONCLUSION Our findings suggest that the P2Y2R represents a novel therapeutic target for the treatment of Sjögren's syndrome.
Collapse
|
44
|
Simeonova D, Stoyanov D, Leunis JC, Murdjeva M, Maes M. Construction of a nitro-oxidative stress-driven, mechanistic model of mood disorders: A nomothetic network approach. Nitric Oxide 2021; 106:45-54. [PMID: 33186727 DOI: 10.1016/j.niox.2020.11.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/05/2020] [Indexed: 12/17/2022]
Abstract
Major depression is accompanied by increased IgM-mediated autoimmune responses to oxidative specific epitopes (OSEs) and nitric oxide (NO)-adducts. These responses were not examined in bipolar disorder type 1 (BP1) and BP2. IgM responses to malondialdehyde (MDA), phosphatidinylinositol, oleic acid, azelaic acid, and NO-adducts were determined in 35 healthy controls, and 47 major depressed (MDD), 29 BP1, and 25 BP2 patients. We also measured serum peroxides, IgG to oxidized LDL (oxLDL), and IgM/IgA directed to lipopolysaccharides (LPS). IgM responses to OSEs and NO-adducts (OSENO) were significantly higher in MDD and BP1 as compared with controls, and IgM to OSEs higher in MDD than in BP2. Partial Least Squares (PLS) analysis showed that 57.7% of the variance in the clinical phenome of mood disorders was explained by number of episodes, a latent vector extracted from IgM to OSENO, IgG to oxLDL, and peroxides. There were significant specific indirect effects of IgA/IgM to LPS on the clinical phenome, which were mediated by peroxides, IgM OSENO, and IgG oxLDL. Using PLS we have constructed a data-driven nomothetic network which ensembled causome (increased plasma LPS load), adverse outcome pathways (namely neuro-affective toxicity), and clinical phenome features of mood disorders in a data-driven model. Based on those feature sets, cluster analysis discovered a new diagnostic class characterized by increased plasma LPS load, peroxides, autoimmune responses to OSENO, and increased phenome scores. Using the new nomothetic network approach, we constructed a mechanistically transdiagnostic diagnostic class indicating neuro-affective toxicity in 74.3% of the mood disorder patients.
Collapse
Affiliation(s)
- Denitsa Simeonova
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University, Plovdiv, Bulgaria
| | - Drozdstoy Stoyanov
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria; Research Institute, Medical University, Plovdiv, Bulgaria
| | | | - Marianna Murdjeva
- Research Institute, Medical University, Plovdiv, Bulgaria; Department of Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria; Section of Immunological Assessment of Chronic Stress, Technological Center of Emergency Medicine, Plovdiv, Bulgaria
| | - Michael Maes
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria; Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Impact Research Center, Deakin University, Geelong, Australia.
| |
Collapse
|
45
|
Matsuda Y, Hiramitsu T, Li XK, Watanabe T. Characteristics of Immunoglobulin M Type Antibodies of Different Origins from the Immunologic and Clinical Viewpoints and Their Application in Controlling Antibody-Mediated Allograft Rejection. Pathogens 2020; 10:pathogens10010004. [PMID: 33374617 PMCID: PMC7822424 DOI: 10.3390/pathogens10010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/25/2022] Open
Abstract
Antibody-mediated allograft rejection (AMR) hinders patient prognosis after organ transplantation. Current studies concerning AMR have mainly focused on the diagnostic value of immunoglobulin G (IgG)-type donor-specific antihuman leukocyte antigen antibodies (DSAs), primarily because of their antigen specificity, whereas the clinical significance of immunoglobulin M (IgM)-type DSAs has not been thoroughly investigated in the context of organ transplantation because of their nonspecificity against antigens. Although consensus regarding the clinical significance and role of IgM antibodies is not clear, as discussed in this review, recent findings strongly suggest that they also have a huge potential in novel diagnostic as well as therapeutic application for the prevention of AMR. Most serum IgM antibodies are known to comprise natural antibodies with low affinity toward antigens, and this is derived from B-1 cells (innate B cells). However, some of the serum IgM-type antibodies reportedly also produced by B-2 cells (conventional B cells). The latter are known to have a high affinity for donor-specific antigens. In this review, we initially discuss how IgM-type antibodies of different origins participate in the pathology of various diseases, directly or through cell surface receptors, complement activation, or cytokine production. Then, we discuss the clinical applicability of B-1 and B-2 cell-derived IgM-type antibodies for controlling AMR with reference to the involvement of IgM antibodies in various pathological conditions.
Collapse
Affiliation(s)
- Yoshiko Matsuda
- Division of Transplant Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan;
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
- Correspondence:
| | - Takahisa Hiramitsu
- Department of Transplant and Endocrine Surgery, Nagoya Daini Red Cross-Hospital, Aichi 466-8650, Japan;
| | - Xiao-kang Li
- Division of Transplant Immunology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan;
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan;
| |
Collapse
|
46
|
Skin-Associated B Cells in the Pathogenesis of Cutaneous Autoimmune Diseases-Implications for Therapeutic Approaches. Cells 2020; 9:cells9122627. [PMID: 33297481 PMCID: PMC7762338 DOI: 10.3390/cells9122627] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
B lymphocytes are crucial mediators of systemic immune responses and are known to be substantial in the pathogenesis of autoimmune diseases with cutaneous manifestations. Amongst them are lupus erythematosus, dermatomyositis, systemic sclerosis and psoriasis, and particularly those driven by autoantibodies such as pemphigus and pemphigoid. However, the concept of autoreactive skin-associated B cells, which may reside in the skin and locally contribute to chronic inflammation, is gradually evolving. These cells are believed to differ from B cells of primary and secondary lymphoid organs and may provide additional features besides autoantibody production, including cytokine expression and crosstalk to autoreactive T cells in an antigen-presenting manner. In chronically inflamed skin, B cells may appear in tertiary lymphoid structures. Those abnormal lymph node-like structures comprise a network of immune and stromal cells possibly enriched by vascular structures and thus constitute an ideal niche for local autoimmune responses. In this review, we describe current considerations of different B cell subsets and their assumed role in skin autoimmunity. Moreover, we discuss traditional and B cell-associated approaches for the treatment of autoimmune skin diseases, including drugs targeting B cells (e.g., CD19- and CD20-antibodies), plasma cells (e.g., proteasome inhibitors, CXCR4 antagonists), activated pathways (such as BTK- and PI3K-inhibitors) and associated activator molecules (BLyS, APRIL).
Collapse
|
47
|
Atosuo J, Karhuvaara O, Suominen E, Vilén L, Nuutila J, Putus T. Indoor-related microbe damage induces complement system activation in building users. Innate Immun 2020; 27:15-22. [PMID: 33287602 PMCID: PMC7780350 DOI: 10.1177/1753425920966641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In this comparative study, serum complement system antimicrobial activity was measured from 159 serum samples, taken from individuals from microbe-damaged (70 samples) and from reference buildings (89 samples). Antimicrobial activity was assessed using a probe-based bacterial Escherichia coli-lux bioluminescence system and comparison was made at a group level between the experimental and reference group. The complement activity was higher in users of microbe-damaged buildings compared with the reference group and the significant (P < 0.001) increase in activity was found in the classical reaction pathway. This study strengthens our notion that exposure to indoor-related microbe damage increases the risk for systemic subclinical inflammation and creates a health risk for building users.
Collapse
Affiliation(s)
- Janne Atosuo
- The Laboratory of Immunochemistry, Department of Biochemistry, Faculty of Science and Engineering, University of Turku, Finland.,Environmental Medicine and Occupational Health, Department of Clinical Medicine, Faculty of Medicine, University of Turku, Finland
| | - Outi Karhuvaara
- The Laboratory of Immunochemistry, Department of Biochemistry, Faculty of Science and Engineering, University of Turku, Finland.,Environmental Medicine and Occupational Health, Department of Clinical Medicine, Faculty of Medicine, University of Turku, Finland
| | - Eetu Suominen
- The Laboratory of Immunochemistry, Department of Biochemistry, Faculty of Science and Engineering, University of Turku, Finland.,Environmental Medicine and Occupational Health, Department of Clinical Medicine, Faculty of Medicine, University of Turku, Finland
| | - Liisa Vilén
- Environmental Medicine and Occupational Health, Department of Clinical Medicine, Faculty of Medicine, University of Turku, Finland
| | - Jari Nuutila
- The Laboratory of Immunochemistry, Department of Biochemistry, Faculty of Science and Engineering, University of Turku, Finland
| | - Tuula Putus
- Environmental Medicine and Occupational Health, Department of Clinical Medicine, Faculty of Medicine, University of Turku, Finland
| |
Collapse
|
48
|
Orwoll ES, Wiedrick J, Nielson CM, Jacobs J, Baker ES, Piehowski P, Petyuk V, Gao Y, Shi T, Smith RD, Bauer DC, Cummings SR, Lapidus J. Proteomic assessment of serum biomarkers of longevity in older men. Aging Cell 2020; 19:e13253. [PMID: 33078901 PMCID: PMC7681066 DOI: 10.1111/acel.13253] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/30/2020] [Accepted: 08/30/2020] [Indexed: 12/28/2022] Open
Abstract
The biological bases of longevity are not well understood, and there are limited biomarkers for the prediction of long life. We used a high-throughput, discovery-based proteomics approach to identify serum peptides and proteins that were associated with the attainment of longevity in a longitudinal study of community-dwelling men age ≥65 years. Baseline serum in 1196 men were analyzed using liquid chromatography-ion mobility-mass spectrometry, and lifespan was determined during ~12 years of follow-up. Men who achieved longevity (≥90% expected survival) were compared to those who died earlier. Rigorous statistical methods that controlled for false positivity were utilized to identify 25 proteins that were associated with longevity. All these proteins were in lower abundance in long-lived men and included a variety involved in inflammation or complement activation. Lower levels of longevity-associated proteins were also associated with better health status, but as time to death shortened, levels of these proteins increased. Pathway analyses implicated a number of compounds as important upstream regulators of the proteins and implicated shared networks that underlie the observed associations with longevity. Overall, these results suggest that complex pathways, prominently including inflammation, are linked to the likelihood of attaining longevity. This work may serve to identify novel biomarkers for longevity and to understand the biology underlying lifespan.
Collapse
Affiliation(s)
| | | | | | - Jon Jacobs
- Biological Science Division Pacific Northwest National Laboratory Richland WA USA
| | - Erin S. Baker
- Department of Chemistry North Carolina State University Raleigh NC USA
| | - Paul Piehowski
- Biological Science Division Pacific Northwest National Laboratory Richland WA USA
| | - Vladislav Petyuk
- Biological Science Division Pacific Northwest National Laboratory Richland WA USA
| | - Yuqian Gao
- Biological Science Division Pacific Northwest National Laboratory Richland WA USA
| | - Tujin Shi
- Biological Science Division Pacific Northwest National Laboratory Richland WA USA
| | - Richard D. Smith
- Biological Science Division Pacific Northwest National Laboratory Richland WA USA
| | - Douglas C. Bauer
- Departments of Medicine and Epidemiology & Biostatistics University of California San Francisco CA USA
| | - Steven R. Cummings
- California Pacific Medical Center Research Institute San Francisco CA USA
| | - Jodi Lapidus
- Oregon Health & Science University Portland OR USA
| | | |
Collapse
|
49
|
Abstract
Coronavirus disease 2019 (COVID-19) is a life-threatening respiratory illness caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Its clinical presentation can vary from the asymptomatic state to acute respiratory distress syndrome (ARDS) and multi-organ dysfunction. Due to our insufficient understanding of its pathophysiology and lack of effective treatment, the morbidity and mortality of severe COVID-19 patients are high. Patients with COVID-19 develop ARDS fueled by exaggerated neutrophil influx into the lungs and cytokine storm. B-1a cells represent a unique subpopulation of B lymphocytes critical for circulating natural antibodies, innate immunity, and immunoregulation. These cells spontaneously produce natural IgM, interleukin (IL)-10, and granulocyte-monocyte colony stimulating factor (GM-CSF). Natural IgM neutralizes viruses and opsonizes bacteria, IL-10 attenuates the cytokine storm, and GM-CSF induces IgM production by B-1a cells in an autocrine manner. Indeed, B-1a cells have been shown to ameliorate influenza virus infection, sepsis, and pneumonia, all of which are similar to COVID-19. The recent discovery of B-1a cells in humans further reinforces their potentially critical role in the immune response against SARS-CoV-2 and their anticipated translational applications against viral and microbial infections. Given that B-1a cells protect against ARDS via immunoglobulin production and the anti-COVID-19 effects of convalescent plasma treatment, we recommend that studies be conducted to further examine the role of B-1a cells in the pathogenesis of COVID-19 and explore their therapeutic potential to treat COVID-19 patients.
Collapse
|
50
|
Reyneveld GIJ, Savelkoul HFJ, Parmentier HK. Current Understanding of Natural Antibodies and Exploring the Possibilities of Modulation Using Veterinary Models. A Review. Front Immunol 2020; 11:2139. [PMID: 33013904 PMCID: PMC7511776 DOI: 10.3389/fimmu.2020.02139] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/06/2020] [Indexed: 12/20/2022] Open
Abstract
Natural antibodies (NAb) are defined as germline encoded immunoglobulins found in individuals without (known) prior antigenic experience. NAb bind exogenous (e.g., bacterial) and self-components and have been found in every vertebrate species tested. NAb likely act as a first-line immune defense against infections. A large part of NAb, so called natural autoantibodies (NAAb) bind to and clear (self) neo-epitopes, apoptotic, and necrotic cells. Such self-binding antibodies cannot, however, be considered as pathogenic autoantibodies in the classical sense. IgM and IgG NAb and NAAb and their implications in health and disease are relatively well-described in humans and mice. NAb are present in veterinary (and wildlife) species, but their relation with diseases and disorders in veterinary species are much less known. Also, there is little known of IgA NAb. IgA is the most abundant immunoglobulin with essential pro-inflammatory and homeostatic properties urging for more research on the importance of IgA NAb. Since NAb in humans were indicated to fulfill important functions in health and disease, their role in health of veterinary species should be investigated more often. Furthermore, it is unknown whether levels of NAb-isotypes and/or idiotypes can and should be modulated. Veterinary species as models of choice fill in a niche between mice and (non-human) primates, and the study of NAb in veterinary species may provide valuable new insights that will likely improve health management. Below, examples of the involvement of NAb in several diseases in mostly humans are shown. Possibilities of intravenous immunoglobulin administration, targeted immunotherapy, immunization, diet, and genetic modulation are discussed, all of which could be well-studied using animal models. Arguments are given why veterinary immunology should obtain inspiration from human studies and why human immunology would benefit from veterinary models. Within the One Health concept, findings from veterinary (and wildlife) studies can be related to human studies and vice versa so that both fields will mutually benefit. This will lead to a better understanding of NAb: their origin, activation mechanisms, and their implications in health and disease, and will lead to novel health management strategies for both human and veterinary species.
Collapse
Affiliation(s)
- G. IJsbrand Reyneveld
- Faculty of Science, VU University, Amsterdam, Netherlands
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Huub F. J. Savelkoul
- Cell Biology and Immunology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Henk K. Parmentier
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| |
Collapse
|