1
|
Liu P, Zuo J, Lu H, Zhang B, Wu C. Bacillus subtilis Fed to Sows Promotes Intestinal Development and Regulates Mucosal Immunity in Offspring. Vet Sci 2025; 12:489. [PMID: 40431582 PMCID: PMC12116194 DOI: 10.3390/vetsci12050489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Diarrhea in piglets causes intestinal inflammation and epithelial damage. Weaned piglets fed with Bacillus subtilis (B.S) have enhanced intestinal mucosal immunity and reduces diarrhea in piglets. However, the immune system of newborn piglets is immature, and B.S cannot effectively activate the intestinal mucosal reaction when given directly. This research explored the impact of the maternal supplementation of B.S-Dia during the final 35 days of gestation on piglet intestinal development and mucosal immunity. The results demonstrated that B.S-Dia administration significantly increased the body weight, jejunal villus height, and crypt depth in the piglets. In addition, B.S-Dia also significantly increased the proliferative activity of intestinal epithelial cells, as evidenced by proliferating cell nuclear antigen (PCNA) staining and the elevated mRNA expression of the proliferation-related gene (c-Myc). Furthermore, B.S-Dia supplementation also reinforced the intestinal mucosal barrier by increasing goblet cell numbers and upregulating the mRNA expression of antimicrobial peptides, such as Muc2 and Lyz-1. Finally, elevated levels of IL-4 and IFN-γ, along with an increased abundance of CD3+ T cells, revealed that the intestinal mucosal immunity of piglets was improved after B.S-Dia administration. Our study indicates that feeding B.S-Dia to sow spromotes intestinal development and improves intestinal mucosal immunity in piglets.
Collapse
Affiliation(s)
- Peng Liu
- College of Veterinary Medicine, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| | - Jinjiao Zuo
- Pet Science and Technology College, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (J.Z.); (B.Z.)
| | - Hui Lu
- College of Veterinary Medicine, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| | - Bin Zhang
- Pet Science and Technology College, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China; (J.Z.); (B.Z.)
| | - Caihong Wu
- College of Veterinary Medicine, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225300, China;
| |
Collapse
|
2
|
Vitari N, Roy S. Intestinal immunoglobulins under microbial dysbiosis: implications in opioid-induced microbial dysbiosis. Front Microbiol 2025; 16:1580661. [PMID: 40297286 PMCID: PMC12034684 DOI: 10.3389/fmicb.2025.1580661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Intestinal immunoglobulins (Igs) maintain homeostasis between the microbiome and host. IgA facilitates microbial balance through a variety of increasingly well-described mechanisms. However, IgM and IgG have less defined intestinal functions but have the potential to activate clearance mechanisms such as the complement system and receptor-mediated bacterial killing. Very little is known regarding the role of Igs under microbial dysbiosis. In this review, we explore how Igs sculpt the intestinal microbiome and respond to microbial dysbiosis. We discuss how IgM, IgA, IgG, and complement individually maintain harmony with the microbiome and consider how these mechanisms could work in synergy. Finally, we explore using an opioid-induced microbial dysbiosis as a model to elucidate immediate changes in Ig-bacterial interactions.
Collapse
Affiliation(s)
- Nicolas Vitari
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sabita Roy
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
3
|
Niu Z, Wang M, Yan Y, Jin X, Ning L, Xu B, Wang Y, Hao Y, Luo Z, Guo C, Zhi L, Zhu W. Challenges in the Development of NK-92 Cells as an Effective Universal Off-the-Shelf Cellular Therapeutic. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1318-1328. [PMID: 39291926 DOI: 10.4049/jimmunol.2400173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024]
Abstract
The human-derived NK-92 cell-based CAR-NK therapy exhibits inconsistency with overall suboptimal efficacy and rapid in vivo clearance of CAR-NK92 cells in cancer patients. Analysis indicates that although pre-existing IgM in healthy individuals (n = 10) strongly recognizes both NK-92 and CAR-NK92 cells, IgG and IgE do not. However, only a subset of cancer patients (3/8) exhibit strong IgM recognition of these cells, with some (2/8) showing pre-existing IgG recognition. These results suggest a natural immunoreactivity between NK-92 and CAR-NK92 cells and pre-existing human Abs. Furthermore, the therapy's immunogenicity is evidenced by enhanced IgG and IgM recognition postinfusion of CAR-NK92 cells. We also confirmed that healthy plasma's cytotoxicity toward these cells is reduced by complement inhibitors, suggesting that Abs may facilitate the rapid clearance of CAR-NK92 cells through complement-dependent cytotoxicity. Given that NK-92 cells lack known receptors for IgG and IgM, identifying and modifying the recognition targets for these Abs on NK-92 and CAR-NK92 cells may improve clinical outcomes. Moreover, we discovered that the 72nd amino acid of the NKG2D receptor on NK-92 cells is alanine. Previous studies have demonstrated polymorphism at the 72nd amino acid of the NKG2D on human NK cells, with NKG2D72Thr exhibiting a superior activation effect on NK cells compared with NKG2D72Ala. We confirmed this conclusion also applies to NK-92 cells by in vitro cytotoxicity experiments. Therefore, reducing the immunoreactivity and immunogenicity of CAR-NK92 and directly switching NK-92 bearing NKG2D72Ala to NKG2D72Thr represent pressing challenges in realizing NK-92 cells as qualified universal off-the-shelf cellular therapeutics.
Collapse
Affiliation(s)
- Zhiyuan Niu
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Mengjun Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yangchun Yan
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xinru Jin
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Linwei Ning
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Bingqian Xu
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yanfeng Wang
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yuekai Hao
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhixia Luo
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Changjiang Guo
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Lingtong Zhi
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Wuling Zhu
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan, China
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
4
|
Wang Y, Xiao J. Recent advances in the molecular understanding of immunoglobulin A. FEBS J 2024; 291:3597-3603. [PMID: 38329005 DOI: 10.1111/febs.17089] [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: 11/29/2023] [Revised: 01/11/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Immunoglobulin A (IgA) plays a crucial role in the human immune system, particularly in mucosal immunity. IgA antibodies that target the mucosal surface are made up of two to five IgA monomers linked together by the joining chain, forming polymeric molecules. These IgA polymers are transported across mucosal epithelial cells by the polymeric immunoglobulin receptor pIgR, resulting in the formation of secretory IgA (SIgA). This review aims to explore recent advancements in our molecular understanding of IgA, with a specific focus on SIgA, and the interaction between IgA and pathogen molecules.
Collapse
Affiliation(s)
- Yuxin Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Junyu Xiao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
5
|
Kumar Bharathkar S, Stadtmueller BM. Structural and Biochemical Requirements for Secretory Component Interactions with Dimeric IgA. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:226-234. [PMID: 38809110 PMCID: PMC11233122 DOI: 10.4049/jimmunol.2300717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Secretory (S) IgA is the predominant mucosal Ab that protects host epithelial barriers and promotes microbial homeostasis. SIgA production occurs when plasma cells assemble two copies of monomeric IgA and one joining chain (JC) to form dimeric (d) IgA, which is bound by the polymeric Ig receptor (pIgR) on the basolateral surface of epithelial cells and transcytosed to the apical surface. There, pIgR is proteolytically cleaved, releasing SIgA, a complex of the dIgA and the pIgR ectodomain, called the secretory component (SC). The pIgR's five Ig-like domains (D1-D5) undergo a conformational change upon binding dIgA, ultimately contacting four IgA H chains and the JC in SIgA. In this study, we report structure-based mutational analysis combined with surface plasmon resonance binding assays that identify key residues in mouse SC D1 and D3 that mediate SC binding to dIgA. Residues in D1 CDR3 are likely to initiate binding, whereas residues that stabilize the D1-D3 interface are likely to promote the conformational change and stabilize the final SIgA structure. Additionally, we find that the JC's three C-terminal residues play a limited role in dIgA assembly but a significant role in pIgR/SC binding to dIgA. Together, these results inform models for the intricate mechanisms underlying IgA transport across epithelia and functions in the mucosa.
Collapse
Affiliation(s)
- Sonya Kumar Bharathkar
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
| | - Beth M. Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Carl R. Woese Institute of Genomic Biology
| |
Collapse
|
6
|
Laprise F, Arduini A, Duguay M, Pan Q, Liang C. SARS-CoV-2 Accessory Protein ORF8 Targets the Dimeric IgA Receptor pIgR. Viruses 2024; 16:1008. [PMID: 39066171 PMCID: PMC11281603 DOI: 10.3390/v16071008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
SARS-CoV-2 is a highly pathogenic respiratory virus that successfully initiates and establishes its infection at the respiratory mucosa. However, little is known about how SARS-CoV-2 antagonizes the host's mucosal immunity. Recent findings have shown a marked reduction in the expression of the polymeric Ig receptor (pIgR) in COVID-19 patients. This receptor maintains mucosal homeostasis by transporting the dimeric IgA (dIgA) and pentameric IgM (pIgM) across mucosal epithelial cells to neutralize the invading respiratory pathogens. By studying the interaction between pIgR and SARS-CoV-2 proteins, we discovered that the viral accessory protein Open Reading Frame 8 (ORF8) potently downregulates pIgR expression and that this downregulation activity of ORF8 correlates with its ability to interact with pIgR. Importantly, the ORF8-mediated downregulation of pIgR diminishes the binding of dIgA or pIgM, and the ORF8 proteins of the variants of concern of SARS-CoV-2 preserve the function of downregulating pIgR, indicating the importance of this conserved activity of ORF8 in SARS-CoV-2 pathogenesis. We further observed that the secreted ORF8 binds to cell surface pIgR, but that this interaction does not trigger the cellular internalization of ORF8, which requires the binding of dIgA to pIgR. These findings suggest the role of ORF8 in SARS-CoV-2 mucosal immune evasion.
Collapse
Affiliation(s)
- Frederique Laprise
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
| | - Ariana Arduini
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| | - Mathew Duguay
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
- Institut de Recherche Clinique de Montréal, Montreal, QC H2W 1R7, Canada
| | - Qinghua Pan
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
| | - Chen Liang
- Lady Davis Institute, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; (F.L.); (A.A.); (M.D.); (Q.P.)
- Department of Microbiology and Immunology, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Medicine, McGill University, Montreal, QC H3G 2M1, Canada
| |
Collapse
|
7
|
Dishaw LJ, Litman GW, Liberti A. Tethering of soluble immune effectors to mucin and chitin reflects a convergent and dynamic role in gut immunity. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230078. [PMID: 38497268 PMCID: PMC10945408 DOI: 10.1098/rstb.2023.0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 12/04/2023] [Indexed: 03/19/2024] Open
Abstract
The immune system employs soluble effectors to shape luminal spaces. Antibodies are soluble molecules that effect immunological responses, including neutralization, opsonization, antibody-dependent cytotoxicity and complement activation. These molecules are comprised of immunoglobulin (Ig) domains. The N-terminal Ig domains recognize antigen, and the C-terminal domains facilitate their elimination through phagocytosis (opsonization). A less-recognized function mediated by the C-terminal Ig domains of the IgG class of antibodies (Fc region) involves the formation of multiple low-affinity bonds with the mucus matrix. This association anchors the antibody molecule to the matrix to entrap potential pathogens. Even though invertebrates are not known to have antibodies, protochordates have a class of secreted molecules containing Ig domains that can bind bacteria and potentially serve a similar purpose. The VCBPs (V region-containing chitin-binding proteins) possess a C-terminal chitin-binding domain that helps tether them to chitin-rich mucus gels, mimicking the IgG-mediated Fc trapping of microbes in mucus. The broad functional similarity of these structurally divergent, Ig-containing, secreted effectors makes a case for a unique form of convergent evolution within chordates. This opinion essay highlights emerging evidence that divergent secreted immune effectors with Ig-like domains evolved to manage immune recognition at mucosal surfaces in strikingly similar ways. This article is part of the theme issue 'Sculpting the microbiome: how host factors determine and respond to microbial colonization'.
Collapse
Affiliation(s)
- L. J. Dishaw
- Morsani College of Medicine, Department of Pediatrics, University of South Florida, Children's Research Institute, St. Petersburg, FL 33701, USA
| | - G. W. Litman
- Morsani College of Medicine, Department of Pediatrics, University of South Florida, Children's Research Institute, St. Petersburg, FL 33701, USA
| | - A. Liberti
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, 80122 Naples, Italy
| |
Collapse
|
8
|
Kumar Bharathkar S, Stadtmueller BM. Structural and biochemical requirements for secretory component interactions with dimeric Immunoglobulin A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.09.566401. [PMID: 38014291 PMCID: PMC10680632 DOI: 10.1101/2023.11.09.566401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Secretory (S) Immunoglobulin (Ig) A is the predominant mucosal antibody that protects host epithelial barriers and promotes microbial homeostasis. SIgA production occurs when plasma cells assemble two copies of monomeric IgA and one joining-chain (JC) to form dimeric (d) IgA, which is bound by the polymeric Ig receptor (pIgR) on the basolateral surface of epithelial cells and transcytosed to the apical surface. There, pIgR is proteolytically cleaved, releasing SIgA, a complex of the dIgA and the pIgR ectodomain, called secretory component (SC). The pIgR's five Ig-like domains (D1-D5) undergo a conformational change upon binding dIgA, ultimately contacting four IgA heavy chains and the JC in SIgA. Here we report structure-based mutational analysis combined with surface plasmon resonance binding assays that identify key residues in mouse SC D1 and D3 that mediate SC binding to dIgA. Residues in D1 CDR3 are likely to initiate binding whereas residues that stabilize the D1-D3 interface are likely to promote the conformation change and stabilize the final SIgA structure. Additionally, we find that the JC's three C-terminal residues play a limited role in dIgA assembly but a significant role in pIgR/SC binding to dIgA. Together results inform new models for the intricate mechanisms underlying IgA transport across epithelia and functions in the mucosa.
Collapse
Affiliation(s)
| | - Beth M. Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Carle R. Woese Institute of Genomic Biology
| |
Collapse
|
9
|
Sinha D, Yaugel-Novoa M, Waeckel L, Paul S, Longet S. Unmasking the potential of secretory IgA and its pivotal role in protection from respiratory viruses. Antiviral Res 2024; 223:105823. [PMID: 38331200 DOI: 10.1016/j.antiviral.2024.105823] [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: 12/11/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
Mucosal immunity has regained its spotlight amidst the ongoing Coronavirus disease 19 (COVID-19) pandemic, with numerous studies highlighting the crucial role of mucosal secretory IgA (SIgA) in protection against Severe acute respiratory syndrome coronavirus-2 or SARS-CoV-2 infections. The observed limitations in the efficacy of currently authorized COVID-19 vaccines in inducing effective mucosal immune responses remind us of the limitations of systemic vaccination in promoting protective mucosal immunity. This resurgence of interest has motivated the development of vaccine platforms capable of enhancing mucosal responses, specifically the SIgA response, and the development of IgA-based therapeutics. Recognizing viral respiratory infections as a global threat, we would like to comprehensively review the existing knowledge on mucosal immunity, with a particular emphasis on SIgA, in the context of SARS-CoV-2, influenza, and Respiratory Syncytial Virus (RSV) infections. This review aims to describe the structural and functional specificities of SIgA, along with its nuanced role in combating influenza, RSV, and SARS-CoV-2 infections. Subsequent sections further elaborate promising vaccine strategies, including mucosal vaccines against Influenza, RSV, and SARS-CoV-2 respiratory viruses, currently undergoing preclinical and clinical development. Additionally, we address the challenges associated with mucosal vaccine development, concluding with a discussion on IgA-based therapeutics as a promising platform for the treatment of viral respiratory infections. This comprehensive review not only synthesizes current insights into mucosal immunity but also identifies critical knowledge gaps, strengthening the way for further advancements in our current understanding and approaches to combat respiratory viral threats.
Collapse
Affiliation(s)
- Divya Sinha
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France
| | - Melyssa Yaugel-Novoa
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France
| | - Louis Waeckel
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France; Immunology Department, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France
| | - Stéphane Paul
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France; Immunology Department, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France; CIC 1408 Inserm Vaccinology, University Hospital of Saint-Etienne, F42055, Saint-Etienne, France.
| | - Stéphanie Longet
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Univ Lyon, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, CIC 1408 Vaccinology, F42023, Saint-Etienne, France.
| |
Collapse
|
10
|
Esmat K, Jamil B, Kheder RK, Kombe Kombe AJ, Zeng W, Ma H, Jin T. Immunoglobulin A response to SARS-CoV-2 infection and immunity. Heliyon 2024; 10:e24031. [PMID: 38230244 PMCID: PMC10789627 DOI: 10.1016/j.heliyon.2024.e24031] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
The novel coronavirus disease (COVID-19) and its infamous "Variants" of the etiological agent termed Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) has proven to be a global health concern. The three antibodies, IgA, IgM, and IgG, perform their dedicated role as main workhorses of the host adaptive immune system in virus neutralization. Immunoglobulin-A (IgA), also known as "Mucosal Immunoglobulin", has been under keen interest throughout the viral infection cycle. Its importance lies because IgA is predominant mucosal antibody and SARS family viruses primarily infect the mucosal surfaces of human respiratory tract. Therefore, IgA can be considered a diagnostic and prognostic marker and an active infection biomarker for SARS CoV-2 infection. Along with molecular analyses, serological tests, including IgA detection tests, are gaining ground in application as an early detectable marker and as a minimally invasive detection strategy. In the current review, it was emphasized the role of IgA response in diagnosis, host defense strategies, treatment, and prevention of SARS-CoV-2 infection. The data analysis was performed through almost 100 published peer-reviewed research reports and comprehended the importance of IgA in antiviral immunity against SARS-CoV-2 and other related respiratory viruses. Taken together, it is concluded that secretory IgA- Abs can serve as a promising detection tool for respiratory viral diagnosis and treatment parallel to IgG-based therapeutics and diagnostics. Vaccine candidates that target and trigger mucosal immune response may also be employed in future dimensions of research against other respiratory viruses.
Collapse
Affiliation(s)
- Khaleqsefat Esmat
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Baban Jamil
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, KRG, Erbil, Iraq
| | - Ramiar Kaml Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania, Sulaymaniyah, Iraq
| | - Arnaud John Kombe Kombe
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Weihong Zeng
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Huan Ma
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| |
Collapse
|
11
|
Mohd Hanafiah K, Hiebert J, Zubach V, Severini A, Anderson DA, Drummer HE. Dimeric immunoglobulin A as a novel diagnostic marker of measles infection. Microbiol Spectr 2024; 12:e0343723. [PMID: 38078716 PMCID: PMC10783017 DOI: 10.1128/spectrum.03437-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/16/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE The world is facing a measles resurgence, and improved diagnostic tests for measles infection are an urgent World Health Organization research priority. Detection of measles-specific immunoglobulin M (IgM) as a standard diagnostic test has low positive predictive value in elimination settings, and there is a need for new biomarkers of measles infection to enable enhanced surveillance and response to outbreaks. We demonstrate the detection of measles-specific dimeric immunoglobulin A (dIgA) in patients with confirmed measles infections using a new indirect enzyme-linked immunosorbent assay protocol that selects for the dIgA fraction from total IgA in the blood. The magnitude of measles-specific dIgA responses showed a low correlation with IgM responses, and our results highlight the potential of dIgA for further development as an alternative and/or complementary biomarker to IgM for serological diagnosis of measles infection.
Collapse
Affiliation(s)
- Khayriyyah Mohd Hanafiah
- Life Sciences, Macfarlane Burnet Institute, Melbourne, Victoria, Australia
- Department of Biology, School of Arts and Sciences, St. John Fisher University, Rochester, New York, USA
| | - Joanne Hiebert
- Viral Exanthemata and STD Section, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Vanessa Zubach
- Viral Exanthemata and STD Section, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Alberto Severini
- Viral Exanthemata and STD Section, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David A. Anderson
- Life Sciences, Macfarlane Burnet Institute, Melbourne, Victoria, Australia
| | - Heidi E. Drummer
- Life Sciences, Macfarlane Burnet Institute, Melbourne, Victoria, Australia
- Department of Microbiology, Monash University, Docklands, Victoria, Australia
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
12
|
Lyu M, Malyutin AG, Stadtmueller BM. The structure of the teleost Immunoglobulin M core provides insights on polymeric antibody evolution, assembly, and function. Nat Commun 2023; 14:7583. [PMID: 37989996 PMCID: PMC10663602 DOI: 10.1038/s41467-023-43240-z] [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: 03/15/2023] [Accepted: 11/04/2023] [Indexed: 11/23/2023] Open
Abstract
Polymeric (p) immunoglobulins (Igs) serve broad functions during vertebrate immune responses. Typically, pIgs contain between two and six Ig monomers, each with two antigen binding fragments and one fragment crystallization (Fc). In addition, many pIgs assemble with a joining-chain (JC); however, the number of monomers and potential to include JC vary with species and heavy chain class. Here, we report the cryo-electron microscopy structure of IgM from a teleost (t) species, which does not encode JC. The structure reveals four tIgM Fcs linked through eight C-terminal tailpieces (Tps), which adopt a single β-sandwich-like domain (Tp assembly) located between two Fcs. Specifically, two of eight heavy chains fold uniquely, resulting in a structure distinct from mammalian IgM, which typically contains five IgM monomers, one JC and a centrally-located Tp assembly. Together with mutational analysis, structural data indicate that pIgs have evolved a range of assembly mechanisms and structures, each likely to support unique antibody effector functions.
Collapse
Affiliation(s)
- Mengfan Lyu
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Andrey G Malyutin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Beckman Institute, California Institute of Technology, Pasadena, CA, 91125, USA
- Takeda Pharmaceuticals, Cambridge, MA, 02139, USA
| | - Beth M Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
13
|
Bharathkar SK, Miller MJ, Stadtmueller BM. Engineered Secretory Immunoglobulin A provides insights on antibody-based effector mechanisms targeting Clostridiodes difficile. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566291. [PMID: 37986930 PMCID: PMC10659285 DOI: 10.1101/2023.11.08.566291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Secretory (S) Immunoglobin (Ig) A is the predominant mucosal antibody, which mediates host interactions with commensal and pathogenic microbes, including Clostridioides difficile. SIgA adopts a polymeric IgA structure that is bound by secretory component (SC). Despite significance, how SIgA supports diverse effector mechanisms is poorly characterized and SIgA-based therapies nonexistent. We engineered chimeric (c) SIgAs, in which we replaced SC domain D2 with a single domain antibody or a monomeric fluorescent protein, allowing us to investigate and enhance SIgA effector mechanisms. cSIgAs exhibited increased neutralization potency against C. difficile toxins, promoted bacterial clumping and cell rupture, and decreased cytotoxicity. cSIgA also allowed us to visualize and/or quantify C. difficile morphological changes and clumping events. Results reveal mechanisms by which SIgA combats C. difficile infection, demonstrate that cSIgA design can modulate these mechanisms, and demonstrate cSIgA's adaptability to modifications that might target a broad range of antigens and effector mechanisms.
Collapse
Affiliation(s)
- Sonya Kumar Bharathkar
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
| | - Michael J. Miller
- Carle R. Woese Institute of Genomic Biology
- Department of food science and Human Nutrition, University of Illinois Urbana-Champaign, Illinois 61801 USA
| | - Beth M. Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Carle R. Woese Institute of Genomic Biology
| |
Collapse
|
14
|
Scheurer S, Junker AC, He C, Schülke S, Toda M. The Role of IgA in the Manifestation and Prevention of Allergic Immune Responses. Curr Allergy Asthma Rep 2023; 23:589-600. [PMID: 37610671 PMCID: PMC10506939 DOI: 10.1007/s11882-023-01105-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2023] [Indexed: 08/24/2023]
Abstract
PURPOSE OF REVIEW Immunoglobulin A (IgA) mediates immune exclusion of antigens in the gut. Notably, IgA plays also a role in the prevention of IgE-mediated allergies and induction of immune tolerance. The present review addresses the role of IgA in the manifestation of IgE-mediated allergies, including allergen-specific immunotherapy (AIT), the regulation of IgA production, and the mechanism of IgA in immune cell activation. RECENT FINDINGS The majority of studies report an association of IgA with the induction of immune tolerance in IgE-mediated allergies. However, reports on the involvement of humoral and mucosal IgA, IgA subtypes, monomeric and polymeric IgA, and the mechanism of IgA-mediated immune cell activation are confounding. Effects by IgA are likely mediated by alteration of microbiota, IgE-blocking capacity, or activation of inhibitory signaling pathways. However, the precise mechanism of IgA-regulation, the contribution of serum and/or mucosal IgA, and IgA1/2 subtypes, on the manifestation of IgE-mediated allergies, and the underlying immune modulatory mechanism are still elusive.
Collapse
Affiliation(s)
- Stephan Scheurer
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany.
| | - Ann-Christine Junker
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany
| | - Chaoqi He
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Stefan Schülke
- Federal Institute for Vaccines and Biomedicines, Molecular Allergology, Paul-Ehrlich-Institut, Paul-Ehrlich Str., 51-58, 63225, Langen, Germany
- Division of Allergology, Paul-Ehrlich-Institut, Federal Institute for Vaccines and Biomedicines, Langen, Germany
| | - Masako Toda
- Laboratory of Food and Biomolecular Science, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
15
|
Hockenberry A, Slack E, Stadtmueller BM. License to Clump: Secretory IgA Structure-Function Relationships Across Scales. Annu Rev Microbiol 2023; 77:645-668. [PMID: 37713459 DOI: 10.1146/annurev-micro-032521-041803] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2023]
Abstract
Secretory antibodies are the only component of our adaptive immune system capable of attacking mucosal pathogens topologically outside of our bodies. All secretory antibody classes are (a) relatively resistant to harsh proteolytic environments and (b) polymeric. Recent elucidation of the structure of secretory IgA (SIgA) has begun to shed light on SIgA functions at the nanoscale. We can now begin to unravel the structure-function relationships of these molecules, for example, by understanding how the bent conformation of SIgA enables robust cross-linking between adjacent growing bacteria. Many mysteries remain, such as the structural basis of protease resistance and the role of noncanonical bacteria-IgA interactions. In this review, we explore the structure-function relationships of IgA from the nano- to the metascale, with a strong focus on how the seemingly banal "license to clump" can have potent effects on bacterial physiology and colonization.
Collapse
Affiliation(s)
- Alyson Hockenberry
- Department of Environmental Microbiology, Swiss Federal Institute of Aquatic Science and Technology (EAWAG), Dübendorf, Switzerland
- Department of Environmental Systems Science (D-USYS), ETH Zürich, Zürich, Switzerland;
| | - Emma Slack
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland;
- Botnar Research Centre for Child Health, Basel, Switzerland
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Beth M Stadtmueller
- Department of Biochemistry, Center for Biophysics and Quantitative Biology, and Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, Illinois, USA;
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois, Urbana, Illinois, USA
| |
Collapse
|
16
|
Lyu M, Malyutin AG, Stadtmueller BM. The structure of the teleost Immunoglobulin M core provides insights on polymeric antibody evolution, assembly, and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534771. [PMID: 37034677 PMCID: PMC10081254 DOI: 10.1101/2023.03.29.534771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Polymeric (p) immunoglobulins (Igs) serve broad functions during vertebrate immune responses. Typically, pIgs contain between two and six Ig monomers, each with two antigen binding fragments and one fragment crystallization (Fc). In addition, many pIgs assemble with a joining-chain (JC); however, the number of monomers and potential to include JC varies with species and heavy chain class. Here, we report the cryo-electron microscopy structure of IgM from a teleost (t) species, which does not encode JC. The structure revealed four tIgM Fcs linked through eight C-terminal tailpieces (Tps), which adopt a single β-sandwich-like domain (Tp assembly) located between two Fcs. Remarkably, two of eight heavy chains fold uniquely, resulting in a structure distinct from mammalian IgM, which typically contains five IgM monomers, one JC and a centrally-located Tp assembly. Together with mutational analysis, structural data indicate that pIgs have evolved a range of assembly mechanisms and structures, each likely to support unique antibody effector functions.
Collapse
Affiliation(s)
- Mengfan Lyu
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
| | - Andrey G. Malyutin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125 USA
- Beckman Institute, California Institute of Technology, Pasadena, CA 91125 USA
- Present address, Takeda Development Center Americas, San Diego, California 92121
| | - Beth M. Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
| |
Collapse
|
17
|
Stosik M, Tokarz-Deptuła B, Deptuła W. Polymeric immunoglobulin receptor (pIgR) in ray-finned fish (Actinopterygii). FISH & SHELLFISH IMMUNOLOGY 2023; 138:108814. [PMID: 37211331 DOI: 10.1016/j.fsi.2023.108814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023]
Affiliation(s)
- Michał Stosik
- Institute of Biological Sciences, Faculty of Biological Sciences University of Zielona Góra, Poland
| | | | - Wiesław Deptuła
- Institute of Veterinary Medicine, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University in Toruń, Poland
| |
Collapse
|
18
|
Boero E, Vezzani G, Micoli F, Pizza M, Rossi O. Functional assays to evaluate antibody-mediated responses against Shigella: a review. Front Cell Infect Microbiol 2023; 13:1171213. [PMID: 37260708 PMCID: PMC10227456 DOI: 10.3389/fcimb.2023.1171213] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/27/2023] [Indexed: 06/02/2023] Open
Abstract
Shigella is a major global pathogen and the etiological agent of shigellosis, a diarrheal disease that primarily affects low- and middle-income countries. Shigellosis is characterized by a complex, multistep pathogenesis during which bacteria use multiple invasion proteins to manipulate and invade the intestinal epithelium. Antibodies, especially against the O-antigen and some invasion proteins, play a protective role as titres against specific antigens inversely correlate with disease severity; however, the context of antibody action during pathogenesis remains to be elucidated, especially with Shigella being mostly an intracellular pathogen. In the absence of a correlate of protection, functional assays rebuilding salient moments of Shigella pathogenesis can improve our understanding of the role of protective antibodies in blocking infection and disease. In vitro assays are important tools to build correlates of protection. Only recently animal models to recapitulate human pathogenesis, often not in full, have been established. This review aims to discuss in vitro assays to evaluate the functionality of anti-Shigella antibodies in polyclonal sera in light of the multistep and multifaced Shigella infection process. Indeed, measurement of antibody level alone may limit the evaluation of full vaccine potential. Serum bactericidal assay (SBA), and other functional assays such as opsonophagocytic killing assays (OPKA), and adhesion/invasion inhibition assays (AIA), are instead physiologically relevant and may provide important information regarding the role played by these effector mechanisms in protective immunity. Ultimately, the review aims at providing scientists in the field with new points of view regarding the significance of functional assays of choice which may be more representative of immune-mediated protection mechanisms.
Collapse
Affiliation(s)
- Elena Boero
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Giacomo Vezzani
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH) S.r.l., Siena, Italy
| |
Collapse
|
19
|
Chen Q, Menon RP, Masino L, Tolar P, Rosenthal PB. Structural basis for Fc receptor recognition of immunoglobulin M. Nat Struct Mol Biol 2023:10.1038/s41594-023-00985-x. [PMID: 37095205 DOI: 10.1038/s41594-023-00985-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/31/2023] [Indexed: 04/26/2023]
Abstract
Immunoglobulin Fc receptors are cell surface transmembrane proteins that bind to the Fc constant region of antibodies and play critical roles in regulating immune responses by activation of immune cells, clearance of immune complexes and regulation of antibody production. FcμR is the immunoglobulin M (IgM) antibody isotype-specific Fc receptor involved in the survival and activation of B cells. Here we reveal eight binding sites for the human FcμR immunoglobulin domain on the IgM pentamer by cryogenic electron microscopy. One of the sites overlaps with the binding site for the polymeric immunoglobulin receptor (pIgR), but a different mode of FcμR binding explains its antibody isotype specificity. Variation in FcμR binding sites and their occupancy reflects the asymmetry of the IgM pentameric core and the versatility of FcμR binding. The complex explains engagement with polymeric serum IgM and the monomeric IgM B-cell receptor (BCR).
Collapse
Affiliation(s)
- Qu Chen
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Rajesh P Menon
- Immune Receptor Activation Laboratory, The Francis Crick Institute, London, UK
| | - Laura Masino
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Pavel Tolar
- Immune Receptor Activation Laboratory, The Francis Crick Institute, London, UK.
- Institute of Immunity and Transplantation, University College London, London, UK.
| | - Peter B Rosenthal
- Structural Biology of Cells and Viruses Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
20
|
Kubagawa H, Clark C, Skopnik CM, Mahmoudi Aliabadi P, Al-Qaisi K, Teuber R, Jani PK, Radbruch A, Melchers F, Engels N, Wienands J. Physiological and Pathophysiological Roles of IgM Fc Receptor (FcµR) Isoforms. Int J Mol Sci 2023; 24:ijms24065728. [PMID: 36982860 PMCID: PMC10058298 DOI: 10.3390/ijms24065728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/14/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
IgM is the first antibody to emerge during phylogeny, ontogeny, and immune responses and serves as a first line of defense. Effector proteins interacting with the Fc portion of IgM, such as complement and its receptors, have been extensively studied for their functions. IgM Fc receptor (FcµR), identified in 2009, is the newest member of the FcR family and is intriguingly expressed by lymphocytes only, suggesting the existence of distinct functions as compared to the FcRs for switched Ig isotypes, which are expressed by various immune and non-hematopoietic cells as central mediators of antibody-triggered responses by coupling the adaptive and innate immune responses. Results from FcµR-deficient mice suggest a regulatory function of FcµR in B cell tolerance, as evidenced by their propensity to produce autoantibodies of both IgM and IgG isotypes. In this article, we discuss conflicting views about the cellular distribution and potential functions of FcµR. The signaling function of the Ig-tail tyrosine-like motif in the FcµR cytoplasmic domain is now formally shown by substitutional experiments with the IgG2 B cell receptor. The potential adaptor protein associating with FcµR and the potential cleavage of its C-terminal cytoplasmic tail after IgM binding are still enigmatic. Critical amino acid residues in the Ig-like domain of FcµR for interacting with the IgM Cµ4 domain and the mode of interaction are now defined by crystallographic and cryo-electron microscopic analyses. Some discrepancies on these interactions are discussed. Finally, elevated levels of a soluble FcµR isoform in serum samples are described as the consequence of persistent B cell receptor stimulation, as seen in chronic lymphocytic leukemia and probably in antibody-mediated autoimmune disorders.
Collapse
Affiliation(s)
| | - Caren Clark
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| | | | | | | | - Ruth Teuber
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | - Peter K Jani
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | | | - Fritz Melchers
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany
| | - Niklas Engels
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| | - Jürgen Wienands
- Institute of Cellular & Molecular Immunology, University Medical Center, 37073 Göttingen, Germany
| |
Collapse
|
21
|
Eroglu A, Al'Abri IS, Kopec RE, Crook N, Bohn T. Carotenoids and Their Health Benefits as Derived via Their Interactions with Gut Microbiota. Adv Nutr 2023; 14:238-255. [PMID: 36775788 DOI: 10.1016/j.advnut.2022.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/21/2022] [Accepted: 10/28/2022] [Indexed: 12/23/2022] Open
Abstract
Carotenoids have been related to a number of health benefits. Their dietary intake and circulating levels have been associated with a reduced incidence of obesity, diabetes, certain types of cancer, and even lower total mortality. Their potential interaction with the gut microbiota (GM) has been generally overlooked but may be of relevance, as carotenoids largely bypass absorption in the small intestine and are passed on to the colon, where they appear to be in part degraded into unknown metabolites. These may include apo-carotenoids that may have biological effects because of higher aqueous solubility and higher electrophilicity that could better target transcription factors, i.e., NF-κB, PPARγ, and RAR/RXRs. If absorbed in the colon, they could have both local and systemic effects. Certain microbes that may be supplemented were also reported to produce carotenoids in the colon. Although some bactericidal aspects of carotenoids have been shown in vitro, a few studies have also demonstrated a prebiotic-like effect, resulting in bacterial shifts with health-associated properties. Also, stimulation of IgA could play a role in this respect. Carotenoids may further contribute to mucosal and gut barrier health, such as stabilizing tight junctions. This review highlights potential gut-related health-beneficial effects of carotenoids and emphasizes the current research gaps regarding carotenoid-GM interactions.
Collapse
Affiliation(s)
- Abdulkerim Eroglu
- Department of Molecular and Structural Biochemistry, College of Agriculture and Life Sciences, North Carolina State University, Raleigh, NC, USA; Plants for Human Health Institute, North Carolina Research Campus, North Carolina State University, Kannapolis, NC, USA.
| | - Ibrahim S Al'Abri
- Department of Chemical and Biomolecular Engineering, College of Engineering, North Carolina State University, Raleigh, NC, USA
| | - Rachel E Kopec
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, USA; Foods for Health Discovery Theme, The Ohio State University, Columbus, OH, USA
| | - Nathan Crook
- Department of Chemical and Biomolecular Engineering, College of Engineering, North Carolina State University, Raleigh, NC, USA
| | - Torsten Bohn
- Nutrition and Health Research Group, Department of Precision Health, Luxembourg Institute of Health, rue 1 A-B, Thomas Edison, L-1445 Strassen, Luxembourg.
| |
Collapse
|
22
|
Yang D, Hu X, Li H, Xu W, Wu T, Chen J. Molecular cloning and characteristic analysis of polymeric immunoglobulin receptor-like (plgRL) in large yellow croaker (Larimichthys crocea). FISH & SHELLFISH IMMUNOLOGY 2023; 132:108503. [PMID: 36581255 DOI: 10.1016/j.fsi.2022.108503] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 06/17/2023]
Abstract
In the present study, the polyimmunoglobulin receptor-like (pIgRL) of large yellow croaker (Larimichthys crocea) was first cloned and characterized. LcpIgRL's full-length cDNA was 1610 bp, encoding 377 amino acids, and the protein's predicted molecular weight was 41.9 kDa, containing two immunoglobulin-like structural domains. The transcript levels of LcpIgRL in different tissues of healthy large yellow croaker were examined by real-time fluorescence quantitative PCR, and the results showed that the gills and head kidney had the highest levels. Within 36 h of the large yellow croaker being infected with Vibrio harveyi, pIgRL mRNA first increased and then decreased in all determined tissues, with the highest expression in the skin and hindgut. Furthermore, a recombinant protein of the extracellular region of LcpIgRL was expressed in E. coli BL21, and a murine rLcpIgRL polyclonal antibody was prepared, which could react specifically with the natural LcpIgRL in skin mucus, but no natural LcpIgRL was detected in serum. Meanwhile, it was found that the rLcpIgRL could bind to the recombinant IgM and the natural IgM, indicating that LcpIgRL could mediate the transport of IgM in mucus. In addition, rLcpIgRL binds to Aeromonas hydrophila and V. harveyi, as well as lipopolysaccharide (LPS) and various saccharides, and reduced binding to bacteria was observed under LPS treatment, suggesting that LcpIgRL can bind to bacteria to prevent infection and that saccharide binding is an important mechanism of interaction between pIgRL and bacteria.
Collapse
Affiliation(s)
- Du Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China; Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiaoman Hu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Hao Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Wenlong Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Ting Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
23
|
Abstract
Single-pass transmembrane receptors (SPTMRs) represent a diverse group of integral membrane proteins that are involved in many essential cellular processes, including signal transduction, cell adhesion, and transmembrane transport of materials. Dysregulation of the SPTMRs is linked with many human diseases. Despite extensive efforts in past decades, the mechanisms of action of the SPTMRs remain incompletely understood. One major hurdle is the lack of structures of the full-length SPTMRs in different functional states. Such structural information is difficult to obtain by traditional structural biology methods such as X-ray crystallography and nuclear magnetic resonance (NMR). The recent rapid development of single-particle cryo-electron microscopy (cryo-EM) has led to an exponential surge in the number of high-resolution structures of integral membrane proteins, including SPTMRs. Cryo-EM structures of SPTMRs solved in the past few years have tremendously improved our understanding of how SPTMRs function. In this review, we will highlight these progresses in the structural studies of SPTMRs by single-particle cryo-EM, analyze important structural details of each protein involved, and discuss their implications on the underlying mechanisms. Finally, we also briefly discuss remaining challenges and exciting opportunities in the field.
Collapse
Affiliation(s)
- Kai Cai
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
| | - Xuewu Zhang
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Departments of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Corresponding Author: Xuewu Zhang, Department of pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| | - Xiao-chen Bai
- Departments of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Departments of Cell Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75231, USA
- Corresponding Author: Xiao-chen Bai, Department of Biophysics, UT Southwestern Medical Center, Dallas, TX 75390, USA;
| |
Collapse
|
24
|
Davis EC, Castagna VP, Sela DA, Hillard MA, Lindberg S, Mantis NJ, Seppo AE, Järvinen KM. Gut microbiome and breast-feeding: Implications for early immune development. J Allergy Clin Immunol 2022; 150:523-534. [PMID: 36075638 PMCID: PMC9463492 DOI: 10.1016/j.jaci.2022.07.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
Establishment of the gut microbiome during early life is a complex process with lasting implications for an individual's health. Several factors influence microbial assembly; however, breast-feeding is recognized as one of the most influential drivers of gut microbiome composition during infancy, with potential implications for function. Differences in gut microbial communities between breast-fed and formula-fed infants have been consistently observed and are hypothesized to partially mediate the relationships between breast-feeding and decreased risk for numerous communicable and noncommunicable diseases in early life. Despite decades of research on the gut microbiome of breast-fed infants, there are large scientific gaps in understanding how human milk has evolved to support microbial and immune development. This review will summarize the evidence on how breast-feeding broadly affects the composition and function of the early-life gut microbiome and discuss mechanisms by which specific human milk components shape intestinal bacterial colonization, succession, and function.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | | | - David A Sela
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Margaret A Hillard
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Samantha Lindberg
- Department of Biomedical Sciences, University of Albany, Rensselaer, NY
| | - Nicholas J Mantis
- Division of Infectious Diseases, New York State Department of Health, Albany, NY
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
25
|
Ramos ACS, Oliveira LM, Santos YLDCO, Dantas MCS, Walker CIB, Faria AMC, Bueno LL, Dolabella SS, Fujiwara RT. The role of IgA in gastrointestinal helminthiasis: A systematic review. Immunol Lett 2022; 249:12-22. [PMID: 36002066 DOI: 10.1016/j.imlet.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/02/2022] [Accepted: 08/19/2022] [Indexed: 11/29/2022]
Abstract
Immunoglobulin-A (IgA) is an important mediator of immunity and has been associated with protection against several pathogens, although its role in gastrointestinal infections remains unclear. Then, the aim of this systematic review was to synthesize qualitative evidence in respect of IgA as mediator of protective immunity against gastrointestinal helminths. Following recommended guidelines, we searched for articles published between January 1990 and October 2019 that evaluated IgA levels and their association with gastrointestinal helminth infections. Twenty-five articles were included after screening 1,546 titles and abstracts, as well as reading in full 52 selected articles. Consistent associations between higher IgA levels and lower parasitological parameters were only found in mice, rats, and sheep. However, the role of IgA in other host species remains uncertain, making it difficult to create a consensus. Therefore, it is too soon to claim that IgA is an effective protective factor against gastrointestinal helminths, and further studies are still needed.
Collapse
Affiliation(s)
- Anne C S Ramos
- Programa de Pós-Graduação em Biologia Parasitária, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil
| | - Luciana M Oliveira
- Programa de Pós-Graduação em Biologia Parasitária, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil; Departamento de Morfologia, Instituto de Ciências Biológicas e da Saúde, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil
| | - Yvanna L D C O Santos
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil
| | - Marlon C S Dantas
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil
| | - Cristiani I B Walker
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil
| | - Ana M C Faria
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brasil
| | - Lílian L Bueno
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brasil
| | - Silvio S Dolabella
- Programa de Pós-Graduação em Biologia Parasitária, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil; Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil.
| | - Ricardo T Fujiwara
- Programa de Pós-Graduação em Biologia Parasitária, Universidade Federal de Sergipe, São Cristóvão 49100-000, SE, Brasil; Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, MG, Brasil.
| |
Collapse
|
26
|
Sheikh-Mohamed S, Isho B, Chao GY, Zuo M, Cohen C, Lustig Y, Nahass GR, Salomon-Shulman RE, Blacker G, Fazel-Zarandi M, Rathod B, Colwill K, Jamal A, Li Z, de Launay KQ, Takaoka A, Garnham-Takaoka J, Patel A, Fahim C, Paterson A, Li AX, Haq N, Barati S, Gilbert L, Green K, Mozafarihashjin M, Samaan P, Budylowski P, Siqueira WL, Mubareka S, Ostrowski M, Rini JM, Rojas OL, Weissman IL, Tal MC, McGeer A, Regev-Yochay G, Straus S, Gingras AC, Gommerman JL. Systemic and mucosal IgA responses are variably induced in response to SARS-CoV-2 mRNA vaccination and are associated with protection against subsequent infection. Mucosal Immunol 2022; 15:799-808. [PMID: 35468942 PMCID: PMC9037584 DOI: 10.1038/s41385-022-00511-0] [Citation(s) in RCA: 180] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/02/2022] [Accepted: 03/23/2022] [Indexed: 02/04/2023]
Abstract
Although SARS-CoV-2 infects the upper respiratory tract, we know little about the amount, type, and kinetics of antibodies (Ab) generated in the oral cavity in response to COVID-19 vaccination. We collected serum and saliva samples from participants receiving two doses of mRNA COVID-19 vaccines and measured the level of anti-SARS-CoV-2 Ab. We detected anti-Spike and anti-Receptor Binding Domain (RBD) IgG and IgA, as well as anti-Spike/RBD associated secretory component in the saliva of most participants after dose 1. Administration of a second dose of mRNA boosted the IgG but not the IgA response, with only 30% of participants remaining positive for IgA at this timepoint. At 6 months post-dose 2, these participants exhibited diminished anti-Spike/RBD IgG levels, although secretory component-associated anti-Spike Ab were more stable. Examining two prospective cohorts we found that participants who experienced breakthrough infections with SARS-CoV-2 variants had lower levels of vaccine-induced serum anti-Spike/RBD IgA at 2-4 weeks post-dose 2 compared to participants who did not experience an infection, whereas IgG levels were comparable between groups. These data suggest that COVID-19 vaccines that elicit a durable IgA response may have utility in preventing infection. Our study finds that a local secretory component-associated IgA response is induced by COVID-19 mRNA vaccination that persists in some, but not all participants. The serum and saliva IgA response modestly correlate at 2-4 weeks post-dose 2. Of note, levels of anti-Spike serum IgA (but not IgG) at this timepoint are lower in participants who subsequently become infected with SARS-CoV-2. As new surges of SARS-CoV-2 variants arise, developing COVID-19 booster shots that provoke high levels of IgA has the potential to reduce person-to-person transmission.
Collapse
Affiliation(s)
| | - Baweleta Isho
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Gary Y.C. Chao
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Michelle Zuo
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Carmit Cohen
- Sheba Medical Center Tel Hashomer, Ramat Gan, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Yaniv Lustig
- Sheba Medical Center Tel Hashomer, Ramat Gan, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel,Central Virology Laboratory, Public Health Services, Ministry of Health, Sheba Medical Center, Tel-Hashomer, Tel-Aviv University, Tel Aviv-Yafo, Israel,Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - George R. Nahass
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA, USA,University of Illinois-Chicago, College of Medicine, Chicago, USA
| | - Rachel E. Salomon-Shulman
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Grace Blacker
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Bhavisha Rathod
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Alainna Jamal
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Zhijie Li
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Keelia Quinn de Launay
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada,Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Alyson Takaoka
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada,Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Julia Garnham-Takaoka
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada,Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Anjali Patel
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada,Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Christine Fahim
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Aimee Paterson
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Angel Xinliu Li
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Nazrana Haq
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Shiva Barati
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Lois Gilbert
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Karen Green
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | | | - Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Patrick Budylowski
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | | - Samira Mubareka
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada,Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Mario Ostrowski
- Department of Immunology, University of Toronto, Toronto, ON, Canada,Department of Medicine, University of Toronto, Toronto, ON, Canada,Keenan Research Centre for Biomedical Science, Toronto, ON, Canada
| | - James M. Rini
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Olga L. Rojas
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Michal Caspi Tal
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Allison McGeer
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Gili Regev-Yochay
- Sheba Medical Center Tel Hashomer, Ramat Gan, Sackler School of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Sharon Straus
- Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, Toronto, ON, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
27
|
Slack E, Diard M. Resistance is futile? Mucosal immune mechanisms in the context of microbial ecology and evolution. Mucosal Immunol 2022; 15:1188-1198. [PMID: 36329192 PMCID: PMC9705250 DOI: 10.1038/s41385-022-00574-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022]
Abstract
In the beginning it was simple: we injected a protein antigen and studied the immune responses against the purified protein. This elegant toolbox uncovered thousands of mechanisms via which immune cells are activated. However, when we consider immune responses against real infectious threats, this elegant simplification misses half of the story: the infectious agents are typically evolving orders-of-magnitude faster than we are. Nowhere is this more pronounced than in the mammalian large intestine. A bacterium representing only 0.1% of the human gut microbiota will have a population size of 109 clones, each actively replicating. Moreover, the evolutionary pressure from other microbes is at least as profound as direct effects of the immune system. Therefore, to really understand intestinal immune mechanisms, we need to understand both the host response and how rapid microbial evolution alters the apparent outcome of the response. In this review we use the examples of intestinal inflammation and secretory immunoglobulin A (SIgA) to highlight what is already known (Fig. 1). Further, we will explore how these interactions can inform immunotherapy and prophylaxis. This has major implications for how we design effective mucosal vaccines against increasingly drug-resistant bacterial pathogens Fig. 1 THE IMMUNE RESPONSE SHAPES THE FITNESS LANDSCAPE IN THE GASTRO-INTESTINAL TRACT.: The red arrows depict possible evolutionary paths of a novel colonizer along adaptive peaks in the intestinal fitness landscapes that change with the status of the host immune system. The flat surfaces represent the non-null fitness baselines (values x or y) at which a bacterium can establish at minimum carrying capacity. a In the healthy gut, metabolic competence, resistance to aggressions by competitors and predators, swift adaptation to rapid fluctuations as well as surviving acidic pH and the flow of the intestinal content, represent potent selective pressures and as many opportunities for bacteria to increase fitness by phenotypic or genetic variations. b When pathogens trigger acute inflammation, bacteria must adapt to iron starvation, killing by immune cells and antimicrobial peptides, and oxidative stress, while new metabolic opportunities emerge. c When high-affinity SIgA are produced against a bacterium, e.g., after oral vaccination, escape of SIgA by altering or losing surface epitopes becomes crucial for maximum fitness. However, escaping polyvalent SIgA responses after vaccination with "evolutionary trap" vaccines leads to evolutionary trade-offs: A fitness maximum is reached in the vaccinated host gut that represents a major disadvantage for transmission into naïve hosts (fitness diminished below x) (d).
Collapse
Affiliation(s)
- Emma Slack
- Laboratory for Mucosal Immunology, Institute for Food, Nutrition and Health, D-HEST, ETH Zürich, Zürich, Switzerland.
- Botnar Research Institute for Child Health, Basel, Switzerland.
| | - Médéric Diard
- Botnar Research Institute for Child Health, Basel, Switzerland.
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
28
|
Transcytosis of IgA Attenuates Salmonella Invasion in Human Enteroids and Intestinal Organoids. Infect Immun 2022; 90:e0004122. [PMID: 35579465 DOI: 10.1128/iai.00041-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Secretory IgA (SIgA) is the most abundant antibody type in intestinal secretions where it contributes to safeguarding the epithelium from invasive pathogens like the Gram-negative bacterium, Salmonella enterica serovar Typhimurium (STm). For example, we recently reported that passive oral administration of the recombinant monoclonal SIgA antibody, Sal4, to mice promotes STm agglutination in the intestinal lumen and restricts bacterial invasion of Peyer's patch tissues. In this report, we sought to recapitulate Sal4-mediated protection against STm in human Enteroids and human intestinal organoids (HIOs) as models to decipher the molecular mechanisms by which antibodies function in mucosal immunity in the human gastrointestinal tract. We confirm that Enteroids and HIO-derived monolayers are permissive to STm infection, dependent on HilD, the master transcriptional regulator of the SPI-I type three secretion system (T3SS). Stimulation of M-like cells in both Enteroids and HIOs by the addition of RANKL further enhanced STm invasion. The apical addition of Sal4 mouse IgA, as well as recombinant human Sal4 dimeric IgA (dIgA) and SIgA resulted a dose-dependent reduction in bacterial invasion. Moreover, basolateral application of Sal4 dIgA to Enteroid and HIO monolayers gave rise to SIgA in the apical compartment via a pathway dependent on expression of the polymeric immunoglobulin receptor (pIgR). The resulting Sal4 SIgA was sufficient to reduce STm invasion of Enteroid and HIO epithelial cell monolayers by ~20-fold. Recombinant Sal4 IgG was also transported in the Enteroid and HIOs, but to a lesser degree and via a pathway dependent on the neonatal Fc receptor (FCGRT). The models described lay the foundation for future studies into detailed mechanisms of IgA and IgG protection against STm and other pathogens.
Collapse
|
29
|
Richards A, Baranova D, Mantis NJ. The prospect of orally administered monoclonal secretory IgA (SIgA) antibodies to prevent enteric bacterial infections. Hum Vaccin Immunother 2022; 18:1964317. [PMID: 34491878 PMCID: PMC9103515 DOI: 10.1080/21645515.2021.1964317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/08/2021] [Accepted: 07/30/2021] [Indexed: 12/20/2022] Open
Abstract
Eliminating diarrheal diseases as a leading cause of childhood morbidity and mortality in low- and middle-income countries (LMICs) will require multiple intervention strategies. In this review, we spotlight a series of preclinical studies investigating the potential of orally administered monoclonal secretory IgA (SIgA) antibodies (MAbs) to reduce disease associated with three enteric bacterial pathogens: Campylobacter jejuni, enterotoxigenic Escherichia coli (ETEC), and invasive Salmonella enterica serovar Typhimurium. IgA MAbs targeting bacterial surface antigens (flagella, adhesins, and lipopolysaccharide) were generated from mice, humanized mice, and human tonsillar B cells. Recombinant SIgA1 and/or SIgA2 derivates of those MAbs were purified from supernatants following transient transfection of 293 cells with plasmids encoding antibody heavy and light chains, J-chain, and secretory component (SC). When administered to mice by gavage immediately prior to (or admixed with) the bacterial challenge, SIgA MAbs reduced infection C. jejuni, ETEC, and S. Typhimurium infections. Fv-matched IgG1 MAbs by comparison were largely ineffective against C. jejuni and S. Typhimurium under the same conditions, although they were partially effective against ETEC. While these findings highlight future applications of orally administered SIgA, the studies also underscored the fundamental challenges associated with using MAbs as prophylactic tools against enteric bacterial diseases.
Collapse
Affiliation(s)
- Angelene Richards
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
| | - Danielle Baranova
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
| | - Nicholas J. Mantis
- Department of Biomedical Sciences, University at Albany School, Albany, NY, USA
- Division of Infectious Diseases, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
30
|
Abstract
As central effectors of the adaptive immune response, immunoglobulins, or antibodies, provide essential protection from pathogens through their ability to recognize foreign antigens, aid in neutralization, and facilitate elimination from the host. Mammalian immunoglobulins can be classified into five isotypes—IgA, IgD, IgE, IgG, and IgM—each with distinct roles in mediating various aspects of the immune response. Of these isotypes, IgA and IgM are the only ones capable of multimerization, arming them with unique biological functions. Increased valency of polymeric IgA and IgM provides high avidity for binding low-affinity antigens, and their ability to be transported across the mucosal epithelium into secretions by the polymeric immunoglobulin receptor allows them to play critical roles in mucosal immunity. Here we discuss the molecular assembly, structure, and function of these multimeric antibodies. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Marissa L. Matsumoto
- Department of Structural Biology, Genentech, Inc., South San Francisco, California, USA
- Current affiliation: Department of Discovery Biotherapeutics, Exelixis, Inc., Alameda, California, USA
| |
Collapse
|
31
|
Mechanism of Neonatal Intestinal Injury Induced by Hyperoxia Therapy. J Immunol Res 2022; 2022:2316368. [PMID: 35071607 PMCID: PMC8769871 DOI: 10.1155/2022/2316368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/16/2021] [Indexed: 11/18/2022] Open
Abstract
High concentration oxygen is widely used in the treatment of neonates, which has a significant effect on improving blood oxygen concentration in neonates with respiratory distress. The adverse effects of hyperoxia therapy on the lung, retina, and neurodevelopment of newborns have been extensively studied, but less attention has been paid to intestinal damage caused by hyperoxia therapy. In this review, we focus on the physical, immune, and microorganism barriers of the intestinal tract and discuss neonatal intestinal tract damage caused by hyperoxia therapy and analyze the molecular mechanism of intestinal damage caused by hyperoxia in combination with necrotizing enterocolitis.
Collapse
|
32
|
3D Structures of IgA, IgM, and Components. Int J Mol Sci 2021; 22:ijms222312776. [PMID: 34884580 PMCID: PMC8657937 DOI: 10.3390/ijms222312776] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 12/02/2022] Open
Abstract
Immunoglobulin G (IgG) is currently the most studied immunoglobin class and is frequently used in antibody therapeutics in which its beneficial effector functions are exploited. IgG is composed of two heavy chains and two light chains, forming the basic antibody monomeric unit. In contrast, immunoglobulin A (IgA) and immunoglobulin M (IgM) are usually assembled into dimers or pentamers with the contribution of joining (J)-chains, which bind to the secretory component (SC) of the polymeric Ig receptor (pIgR) and are transported to the mucosal surface. IgA and IgM play a pivotal role in various immune responses, especially in mucosal immunity. Due to their structural complexity, 3D structural study of these molecules at atomic scale has been slow. With the emergence of cryo-EM and X-ray crystallographic techniques and the growing interest in the structure-function relationships of IgA and IgM, atomic-scale structural information on IgA-Fc and IgM-Fc has been accumulating. Here, we examine the 3D structures of IgA and IgM, including the J-chain and SC. Disulfide bridging and N-glycosylation on these molecules are also summarized. With the increasing information of structure–function relationships, IgA- and IgM-based monoclonal antibodies will be an effective option in the therapeutic field.
Collapse
|
33
|
Abokor AA, McDaniel GH, Golonka RM, Campbell C, Brahmandam S, Yeoh BS, Joe B, Vijay-Kumar M, Saha P. Immunoglobulin A, an Active Liaison for Host-Microbiota Homeostasis. Microorganisms 2021; 9:2117. [PMID: 34683438 PMCID: PMC8539215 DOI: 10.3390/microorganisms9102117] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
Mucosal surfaces in the gastrointestinal tract are continually exposed to native, commensal antigens and susceptible to foreign, infectious antigens. Immunoglobulin A (IgA) provides dual humoral responses that create a symbiotic environment for the resident gut microbiota and prevent the invasion of enteric pathogens. This review features recent immunological and microbial studies that elucidate the underlying IgA and microbiota-dependent mechanisms for mutualism at physiological conditions. IgA derailment and concurrent microbiota instability in pathological diseases are also discussed in detail. Highlights of this review underscore that the source of IgA and its structural form can dictate microbiota reactivity to sustain a diverse niche where both host and bacteria benefit. Other important studies emphasize IgA insufficiency can result in the bloom of opportunistic pathogens that encroach the intestinal epithelia and disseminate into circulation. The continual growth of knowledge in these subjects can lead to the development of therapeutics targeting IgA and/or the microbiota to treat life threatening diseases.
Collapse
Affiliation(s)
- Ahmed A. Abokor
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Grant H. McDaniel
- College of Medicine, University of Toledo, Toledo, OH 43614, USA; (G.H.M.); (C.C.); (S.B.)
| | - Rachel M. Golonka
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Connor Campbell
- College of Medicine, University of Toledo, Toledo, OH 43614, USA; (G.H.M.); (C.C.); (S.B.)
| | - Sreya Brahmandam
- College of Medicine, University of Toledo, Toledo, OH 43614, USA; (G.H.M.); (C.C.); (S.B.)
| | - Beng San Yeoh
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Bina Joe
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Matam Vijay-Kumar
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| | - Piu Saha
- Department of Physiology & Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (A.A.A.); (R.M.G.); (B.S.Y.); (B.J.); (M.V.-K.)
| |
Collapse
|
34
|
Flowers EM, Neely HR, Guo J, Almeida T, Ohta Y, Castro CD, Flajnik MF. Identification of the Fc-alpha/mu receptor in Xenopus provides insight into the emergence of the poly-Ig receptor (pIgR) and mucosal Ig transport. Eur J Immunol 2021; 51:2590-2606. [PMID: 34411303 DOI: 10.1002/eji.202149383] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/05/2021] [Indexed: 01/17/2023]
Abstract
The polyimmunoglobulin receptor (pIgR) transcytoses J chain-containing antibodies through mucosal epithelia. In mammals, two cis-duplicates of PIGR, FCMR, and FCAMR, flank the PIGR gene. A PIGR duplication is first found in amphibians, previously annotated as PIGR2 (herein xlFCAMR), and is expressed by APCs. We demonstrate that xlFcamR is the equivalent of mammalian FcamR. It has been assumed that pIgR is the oldest member of this family, yet our data could not distinguish whether PIGR or FCAMR emerged first; however, FCMR was the last family member to emerge. Interestingly, bony fish "pIgR" is not an orthologue of tetrapod pIgR, and possibly acquired its function via convergent evolution. PIGR/FCAMR/FCMR are members of a larger superfamily, including TREM, CD300, and NKp44, which we name the "double-disulfide Ig superfamily" (ddIgSF). Domains related to each ddIgSF family were identified in cartilaginous fish (sharks, chimeras) and encoded in a single gene cluster syntenic to the human pIgR locus. Thus, the ddIgSF families date back to the earliest antibody-based adaptive immunity, but apparently not before. Finally, our data strongly suggest that the J chain arose in evolution only for Ig multimerization. This study provides a framework for further studies of pIgR and the ddIgSF in vertebrates.
Collapse
Affiliation(s)
- Emily M Flowers
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Harold R Neely
- Department of Immunobiology, Harvard Medical School, Boston, MA, USA
| | - Jacqueline Guo
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tereza Almeida
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yuko Ohta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Caitlin D Castro
- Committee on Immunology and Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL, USA
| | - Martin F Flajnik
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
35
|
Xu J, Wu Y, Xu C, Munang'andu HM, Xu H. Characterization of the Pelodiscus sinensis polymeric immunoglobulin receptor (P. sinensis pIgR) and its response to LPS and Aeromonas sobria. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 121:104072. [PMID: 33798618 DOI: 10.1016/j.dci.2021.104072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 03/12/2021] [Accepted: 03/12/2021] [Indexed: 06/12/2023]
Abstract
The polymeric immunoglobulin receptor (pIgR) is one of the most vital components of mucosal immunity that plays a pivotal role in mediating transcytosis of polymeric immunoglobulin (pIg) on epithelial surfaces for protection against invading pathogens. Herein, we cloned the full-length cDNA of Pelodiscus sinensis pIgR, designated as P. sinensis pIgR, made of an open reading frame (ORF) of 1848 bp, molecular weight of 68.2 kDa and estimated isoelectric point of 7.00. The deduced P. sinensis pIgR sequence had a leader peptide, extracellular region containing four immunoglobulin-like domains (Ig like domains), transmembrane and intracellular regions comparable with other vertebrates. P. sinensis pIgR contained four Ig like domains that corresponded with mammalian D1, D3, D4 and D5 similar with reptile and avian Ig like domains. It had 40 potential phosphorylation sites, four putative N-glycosylation sites and several motifs resembling mammalian pIgR motifs. Phylogenetic analysis showed a close relationship between P. sinensis pIgR with avian and reptile pIgRs. P. sinensis pIgR basal levels were higher in the esophagus, small intestine and intestinnum crissum than in other organs of health turtles. Intragastric delivery of LPS and Aeromonassobria led to significant upregulation of P. sinensis pIgR in tissues of the gastrointestinal tract. A polyclonal anti- P. sinensis pIgR antibody produced in rabbit reacted with the recombinant P. sinensis pIgR protein expressed in Escherichia coli in Western blot. These studies demonstrate the existence and immune response of P. sinensis pIgR to stimulation in mucosal organs in Chinese soft-shelled turtles.
Collapse
Affiliation(s)
- Jiehao Xu
- College of Biological and Environmental Science, Zhejiang Wanli University, Ningbo, 315100, Zhejiang, People's Republic of China
| | - Yue Wu
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Cheng Xu
- Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, PO Box 369, 0102, Oslo, Norway
| | - Hetron Mweemba Munang'andu
- Department of Production Medicine, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, PO Box 369, 0102, Oslo, Norway.
| | - Haisheng Xu
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang Province, People's Republic of China.
| |
Collapse
|
36
|
Kubagawa H, Skopnik CM, Al-Qaisi K, Calvert RA, Honjo K, Kubagawa Y, Teuber R, Aliabadi PM, Enghard P, Radbruch A, Sutton BJ. Differences between Human and Mouse IgM Fc Receptor (FcµR). Int J Mol Sci 2021; 22:ijms22137024. [PMID: 34209905 PMCID: PMC8267714 DOI: 10.3390/ijms22137024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 01/02/2023] Open
Abstract
Both non-immune "natural" and antigen-induced "immune" IgM are important for protection against pathogens and for regulation of immune responses to self-antigens. Since the bona fide IgM Fc receptor (FcµR) was identified in humans by a functional cloning strategy in 2009, the roles of FcµR in these IgM effector functions have begun to be explored. In this short essay, we describe the differences between human and mouse FcµRs in terms of their identification processes, cellular distributions and ligand binding activities with emphasis on our recent findings from the mutational analysis of human FcµR. We have identified at least three sites of human FcµR, i.e., Asn66 in the CDR2, Lys79 to Arg83 in the DE loop and Asn109 in the CDR3, responsible for its constitutive IgM-ligand binding. Results of computational structural modeling analysis are consistent with these mutational data and a model of the ligand binding, Ig-like domain of human FcµR is proposed. Serendipitously, substitution of Glu41 and Met42 in the CDR1 of human FcµR with mouse equivalents Gln and Leu, either single or more prominently in combination, enhances both the receptor expression and IgM binding. These findings would help in the future development of preventive and therapeutic interventions targeting FcµR.
Collapse
Affiliation(s)
- Hiromi Kubagawa
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
- Correspondence: ; Tel.: +49-030-2846-0782
| | - Christopher M. Skopnik
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Khlowd Al-Qaisi
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Rosaleen A. Calvert
- Randall Centre for Cell and Molecular Biophysics, King’s College, London SE1 1UL, UK; (R.A.C.); (B.J.S.)
| | - Kazuhito Honjo
- Department of Pathology of University of Alabama at Birmingham, Birmingham, AL 35294, USA.; (K.H.); (Y.K.)
| | - Yoshiki Kubagawa
- Department of Pathology of University of Alabama at Birmingham, Birmingham, AL 35294, USA.; (K.H.); (Y.K.)
| | - Ruth Teuber
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Pedram Mahmoudi Aliabadi
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Philipp Enghard
- Department of Nephrology and Medical Intensive Care, Charité-Universitätmedizin, 10117 Berlin, Germany;
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum, 10117 Berlin, Germany; (C.M.S.); (K.A.-Q.); (R.T.); (P.M.A.); (A.R.)
| | - Brian J. Sutton
- Randall Centre for Cell and Molecular Biophysics, King’s College, London SE1 1UL, UK; (R.A.C.); (B.J.S.)
| |
Collapse
|
37
|
Richards A, Baranova DE, Pizzuto MS, Jaconi S, Willsey GG, Torres-Velez FJ, Doering JE, Benigni F, Corti D, Mantis NJ. Recombinant Human Secretory IgA Induces Salmonella Typhimurium Agglutination and Limits Bacterial Invasion into Gut-Associated Lymphoid Tissues. ACS Infect Dis 2021; 7:1221-1235. [PMID: 33728898 PMCID: PMC8154420 DOI: 10.1021/acsinfecdis.0c00842] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Indexed: 12/11/2022]
Abstract
As the predominant antibody type in mucosal secretions, human colostrum, and breast milk, secretory IgA (SIgA) plays a central role in safeguarding the intestinal epithelium of newborns from invasive enteric pathogens like the Gram-negative bacterium Salmonella enterica serovar Typhimurium (STm). SIgA is a complex molecule, consisting of an assemblage of two or more IgA monomers, joining (J)-chain, and secretory component (SC), whose exact functions in neutralizing pathogens are only beginning to be elucidated. In this study, we produced and characterized a recombinant human SIgA variant of Sal4, a well-characterized monoclonal antibody (mAb) specific for the O5-antigen of STm lipopolysaccharide (LPS). We demonstrate by flow cytometry, light microscopy, and fluorescence microscopy that Sal4 SIgA promotes the formation of large, densely packed bacterial aggregates in vitro. In a mouse model, passive oral administration of Sal4 SIgA was sufficient to entrap STm within the intestinal lumen and reduce bacterial invasion into gut-associated lymphoid tissues by several orders of magnitude. Bacterial aggregates induced by Sal4 SIgA treatment in the intestinal lumen were recalcitrant to immunohistochemical staining, suggesting the bacteria were encased in a protective capsule. Indeed, a crystal violet staining assay demonstrated that STm secretes an extracellular matrix enriched in cellulose following even short exposures to Sal4 SIgA. Collectively, these results demonstrate that recombinant human SIgA recapitulates key biological activities associated with mucosal immunity and raises the prospect of oral passive immunization to combat enteric diseases.
Collapse
Affiliation(s)
- Angelene
F. Richards
- Department
of Biomedical Sciences, University at Albany
School of Public Health, Albany, New York 12208, United States
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Danielle E. Baranova
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Matteo S. Pizzuto
- Humabs
BioMed SA a Subsidiary of Vir Biotechnology Inc., 6500 Bellinzona, Switzerland
| | - Stefano Jaconi
- Humabs
BioMed SA a Subsidiary of Vir Biotechnology Inc., 6500 Bellinzona, Switzerland
| | - Graham G. Willsey
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Fernando J. Torres-Velez
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Jennifer E. Doering
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| | - Fabio Benigni
- Humabs
BioMed SA a Subsidiary of Vir Biotechnology Inc., 6500 Bellinzona, Switzerland
| | - Davide Corti
- Humabs
BioMed SA a Subsidiary of Vir Biotechnology Inc., 6500 Bellinzona, Switzerland
| | - Nicholas J. Mantis
- Department
of Biomedical Sciences, University at Albany
School of Public Health, Albany, New York 12208, United States
- Division
of Infectious Diseases, Wadsworth Center,
New York State Department of Health, Albany, New York 12208, United States
| |
Collapse
|
38
|
Sheng X, Guo Y, Tang Q, Tang X, Xing J, Chi H, Zhan W. Upregulation of polymeric immunoglobulin receptor expression in flounder (Paralichthys olivaceus) gill cells by cytokine tumor necrosis factor-α via activating PI3K and NF-κB signaling pathways. Mol Immunol 2021; 135:170-182. [PMID: 33901762 DOI: 10.1016/j.molimm.2021.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 04/04/2021] [Accepted: 04/12/2021] [Indexed: 01/04/2023]
Abstract
The polymeric immunoglobulin receptor (pIgR) transports secretory immunoglobulins across mucosal epithelial cells into external secretions, playing critical roles in mucosal surface defenses, but the regulation mechanism of pIgR expression is not clarified in teleost fish. In this study, the dynamic changes of flounder (Paralichthys olivaceus) pIgR (fpIgR) and pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) mRNA expression in mucosal tissues were first analyzed post inactivated Vibrio anguillarum immunization, and increased production of TNF-α was found to correlate with increased expression of fpIgR. To determine that cytokine TNF-α influenced fpIgR expression, following confirming that natural fpIgR expressed on flounder gill (FG) cells, FG cells were incubated with various concentrations of recombinant TNF-α for different time, the results showed that the expressions of fpIgR were significantly upregulated at gene and protein levels in a dose-dependent and time-dependent manner, and similar change trend was observed for free secretory component (SC) secreted by fpIgR into the culture supernatant. After FG cells were treated with TNF-α, specific phosphoinositide 3-kinase (PI3K) inhibitor wortmannin, nuclear factor kappa-B (NF-κB) inhibitor Bay11-7082, and the mixtures of TNF-α and wortmannin / Bay11-7082 respectively, the fpIgR protein and mRNA levels, together with SC secretion, obviously decreased in wortmannin- and Bay11-7082-treated cells compared with the untreated control, and cotreatment with wortmannin / Bay11-7082 plus TNF-α resulted in lower expression compared with that upon treatment with TNF-α alone, indicating that the inhibition of PI3K and NF-κB both blocked the ability of TNF-α to increase cellular fpIgR and SC levels. Furthermore, the gene expressions of PI3K and NF-κB were upregulated and present a tendency to increase first and then decrease after TNF-α treatment of FG cells; However, the expression of PI3K mRNA was inhibited significantly by wortmannin but not by Bay11-7082, and the expression of NF-κB mRNA was suppressed obviously by Bay11-7082 but not by wortmannin, suggesting that inhibition of PI3K or NF-κB had no influence on each other. All these results collectively revealed that TNF-α could transcriptionally upregulate fpIgR expression and SC production, and this TNF-α-induced pIgR expression was regulated by complex mechanisms that involved PI3K and NF-κB signaling pathways, which provided evidences for pro-inflammatory cytokine TNF-α acting as a regulator in pIgR expression and better understanding of regulation mechanism of pIgR expression in teleost fish.
Collapse
Affiliation(s)
- Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, PR China
| | - Yuan Guo
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, PR China
| | - Qian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, PR China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, PR China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, PR China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, PR China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, 266003, PR China; Function Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, PR China.
| |
Collapse
|
39
|
Kabbert J, Benckert J, Rollenske T, Hitch TCA, Clavel T, Cerovic V, Wardemann H, Pabst O. High microbiota reactivity of adult human intestinal IgA requires somatic mutations. J Exp Med 2021; 217:151927. [PMID: 32640466 PMCID: PMC7526496 DOI: 10.1084/jem.20200275] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/25/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
The gut is home to the body’s largest population of plasma cells. In healthy individuals, IgA is the dominating isotype, whereas patients with inflammatory bowel disease also produce high concentrations of IgG. In the gut lumen, secretory IgA binds pathogens and toxins but also the microbiota. However, the antigen specificity of IgA and IgG for the microbiota and underlying mechanisms of antibody binding to bacteria are largely unknown. Here we show that microbiota binding is a defining property of human intestinal antibodies in both healthy and inflamed gut. Some bacterial taxa were commonly targeted by different monoclonal antibodies, whereas others selectively bound single antibodies. Interestingly, individual human monoclonal antibodies from both healthy and inflamed intestines bound phylogenetically unrelated bacterial species. This microbiota cross-species reactivity did not correlate with antibody polyreactivity but was crucially dependent on the accumulation of somatic mutations. Therefore, our data suggest that a system of affinity-matured, microbiota cross-species–reactive IgA is a common aspect of SIgA–microbiota interactions in the gut.
Collapse
Affiliation(s)
- Johanna Kabbert
- Institute of Molecular Medicine, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Julia Benckert
- Max Planck Research Group Molecular Immunology, Max Planck Institute for Infection Biology, Berlin, Germany.,Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Tim Rollenske
- B Cell Immunology, German Cancer Research Centre, Heidelberg, Germany
| | - Thomas C A Hitch
- Functional Microbiome Research Group, Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Vuk Cerovic
- Institute of Molecular Medicine, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| | - Hedda Wardemann
- B Cell Immunology, German Cancer Research Centre, Heidelberg, Germany
| | - Oliver Pabst
- Institute of Molecular Medicine, Rheinisch-Westfälische Technische Hochschule Aachen University, Aachen, Germany
| |
Collapse
|
40
|
Atyeo C, Alter G. The multifaceted roles of breast milk antibodies. Cell 2021; 184:1486-1499. [PMID: 33740451 DOI: 10.1016/j.cell.2021.02.031] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/07/2021] [Accepted: 02/12/2021] [Indexed: 12/20/2022]
Abstract
Neonates are born with an immature immune system and rely on the transfer of immunity from their mothers. Maternal antibodies are transferred via the placenta and breast milk. Although the role of placentally transferred immunoglobulin G (IgG) is established, less is known about the selection of antibodies transferred via breast milk and the mechanisms by which they provide protection against neonatal disease. Evidence suggests that breast milk antibodies play multifaceted roles, preventing infection and supporting the selection of commensals and tolerizing immunity during infancy. Here, we discuss emerging data related to the importance of breast milk antibodies in neonatal immunity and development.
Collapse
Affiliation(s)
- Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; PhD Program in Virology, Division of Medical Sciences, Harvard University, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
41
|
Wei H, Wang JY. Role of Polymeric Immunoglobulin Receptor in IgA and IgM Transcytosis. Int J Mol Sci 2021; 22:ijms22052284. [PMID: 33668983 PMCID: PMC7956327 DOI: 10.3390/ijms22052284] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Transcytosis of polymeric IgA and IgM from the basolateral surface to the apical side of the epithelium and subsequent secretion into mucosal fluids are mediated by the polymeric immunoglobulin receptor (pIgR). Secreted IgA and IgM have vital roles in mucosal immunity in response to pathogenic infections. Binding and recognition of polymeric IgA and IgM by pIgR require the joining chain (J chain), a small protein essential in the formation and stabilization of polymeric Ig structures. Recent studies have identified marginal zone B and B1 cell-specific protein (MZB1) as a novel regulator of polymeric IgA and IgM formation. MZB1 might facilitate IgA and IgM transcytosis by promoting the binding of J chain to Ig. In this review, we discuss the roles of pIgR in transcytosis of IgA and IgM, the roles of J chain in the formation of polymeric IgA and IgM and recognition by pIgR, and focus particularly on recent progress in understanding the roles of MZB1, a molecular chaperone protein.
Collapse
Affiliation(s)
- Hao Wei
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China;
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China;
- Department of Clinical Immunology, Children’s Hospital of Fudan University, Shanghai 201102, China
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
- Correspondence: ; Tel.: +86-(21)-54237957
| |
Collapse
|
42
|
Yang S, Yuan X, Kang T, Xia Y, Xu S, Zhang X, Chen W, Jin Z, Ma Y, Ye Z, Qian S, Huang M, Lv Z, Fei H. Molecular cloning and binding analysis of polymeric immunoglobulin receptor in largemouth bass (Micropterus salmoides). Mol Immunol 2021; 133:14-22. [PMID: 33610122 DOI: 10.1016/j.molimm.2021.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/30/2021] [Accepted: 02/04/2021] [Indexed: 10/22/2022]
Abstract
The polymeric immunoglobulin receptor (pIgR) is an important molecule in the mucosal immunity of teleosts. Previous studies have shown that pIgR can bind and transport polymeric immunoglobulins (pIgs), but few studies have focused on the binding of teleost pIgR to bacteria. In this study, we identified a gene encoding pIgR in largemouth bass (Micropterus salmoides). The pIgR gene contained two Ig-like domains (ILDs), which were homologous to ILD1 and ILD5 of mammalian pIgR. Our results showed that largemouth bass pIgR-ILD could combine with IgM. Moreover, we also found that largemouth bass pIgR-ILD could bind to Aeromonas hydrophila and Micrococcus luteus. Further analysis showed that largemouth bass pIgR-ILD could also combine with lipopolysaccharide (LPS), peptidoglycan (PGN) and various saccharides, and reduced binding to bacteria was observed with LPS and PGN treatment, indicating that largemouth bass pIgR could bind to bacteria to prevent infection and that saccharide binding is an important interaction mechanism between pIgR and bacteria. These results collectively demonstrated that largemouth bass pIgR not only combines with IgM but also binds to bacteria by various saccharides.
Collapse
Affiliation(s)
- Shun Yang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Xiangyu Yuan
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Ting Kang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Yanting Xia
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Shuqi Xu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Xintang Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Wenqi Chen
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Zhihong Jin
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Yuanxin Ma
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Zifeng Ye
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China
| | - Shichao Qian
- Huzhou Baijiayu Biotech Co., Ltd., 313000 Huzhou, China
| | - Mengmeng Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zhengbing Lv
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Hui Fei
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, 310018, China; Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
43
|
Maruthachalam BV, Zwolak A, Lin-Schmidt X, Keough E, Tamot N, Venkataramani S, Geist B, Singh S, Ganesan R. Discovery and characterization of single-domain antibodies for polymeric Ig receptor-mediated mucosal delivery of biologics. MAbs 2021; 12:1708030. [PMID: 31906797 PMCID: PMC6973331 DOI: 10.1080/19420862.2019.1708030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mucosal immunity is dominated by secretory IgA and IgM, although these are less favorable compared to IgG molecules for therapeutic development. Polymeric IgA and IgM are actively transported across the epithelial barrier via engagement of the polymeric Ig receptor (pIgR), but IgG molecules lack a lumen-targeted active transport mechanism, resulting in poor biodistribution of IgG therapeutics in mucosal tissues. In this work, we describe the discovery and characterization of single-domain antibodies (VHH) that engage pIgR and undergo transepithelial transport across the mucosal epithelium. The anti-pIgR VHH panel displayed a broad range of biophysical characteristics, epitope diversity, IgA competition profiles and transcytosis activity in cell and human primary lung tissue models. Making use of this diverse VHH panel, we studied the relationship between biophysical and functional properties of anti-pIgR binders targeting different domains and epitopes of pIgR. These VHH molecules will serve as excellent tools for studying pIgR-mediated transport of biologics and for delivering multispecific IgG antibodies into mucosal lumen, where they can target and neutralize mucosal antigens.
Collapse
Affiliation(s)
| | - Adam Zwolak
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Xiefan Lin-Schmidt
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Edward Keough
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Ninkka Tamot
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Sathya Venkataramani
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Brian Geist
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Sanjaya Singh
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| | - Rajkumar Ganesan
- Janssen BioTherapeutics, Janssen Research and Development, Spring House, PA, USA
| |
Collapse
|
44
|
Kumar N, Arthur CP, Ciferri C, Matsumoto ML. Structure of the human secretory immunoglobulin M core. Structure 2021; 29:564-571.e3. [PMID: 33513362 DOI: 10.1016/j.str.2021.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/22/2020] [Accepted: 01/08/2021] [Indexed: 11/28/2022]
Abstract
Immunoglobulins (Ig) A and M are the only human antibodies that form oligomers and undergo transcytosis to mucosal secretions via the polymeric Ig receptor (pIgR). When complexed with the J-chain (JC) and the secretory component (SC) of pIgR, secretory IgA and IgM (sIgA and sIgM) play critical roles in host-pathogen defense. Recently, we determined the structure of sIgA-Fc which elucidated the mechanism of polymeric IgA assembly and revealed an extensive binding interface between IgA-Fc, JC, and SC. Despite low sequence identity shared with IgA-Fc, IgM-Fc also undergoes JC-mediated assembly and binds pIgR. Here, we report the structure of sIgM-Fc and carryout a systematic comparison to sIgA-Fc. Our structural analysis reveals a remarkably conserved mechanism of JC-templated oligomerization and SC recognition of both IgM and IgA through a highly conserved network of interactions. These studies reveal the structurally conserved features of sIgM and sIgA required for function in mucosal immunity.
Collapse
Affiliation(s)
- Nikit Kumar
- Department of Structural Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Christopher P Arthur
- Department of Structural Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Claudio Ciferri
- Department of Structural Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Marissa L Matsumoto
- Department of Structural Biology, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
45
|
Singh VK, Kumar S, Dhaked RK, Ansari AS, Lohiya NK, Tapryal S. Generation of oligomers of subunit vaccine candidate glycoprotein D of Herpes Simplex Virus-2 expressed in fusion with IgM Fc domain(s) in Escherichia coli: A strategy to enhance the immunogenicity of the antigen. 3 Biotech 2020; 10:463. [PMID: 33047090 PMCID: PMC7541101 DOI: 10.1007/s13205-020-02452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/23/2020] [Indexed: 11/25/2022] Open
Abstract
Glycoprotein D (gD) of Herpes Simplex Virus-2 is used as an antigen in various anti-herpes subunit vaccines owing to its involvement in binding the host cell receptors for host infectivity. However, most of these monomeric protein based candidates have shown low immunogenicity in animal models. To enhance the immunogenicity of gD, a fresh approach of fusing its ectodomain with the Fc domain(s) of IgM has been adopted to oligomerize the viral antigen and to exploite the immune-modulating potential of IgM Fc. Six vaccine constructs, generated by fusing three gD-ectodomain-length-variants with the Ig µ-chain domain 4 (µCH4) and µCH3-CH4 fragment, were cloned in Escherichia coli using pET28b( +) vector. The vaccine proteins were expressed in the form of inclusion bodies (IBs) and were in vitro refolded into protein oligomers of high stoichiometries of ~ 15–24, with 70–80% refolding yields. The conformations of gD and Fc components of the refolded oligomers were analyzed by ELISA and CD spectroscopy and were found to be native-like. The sizes and profiles of the size-distribution of oligomers were determined by dynamic light scattering (DLS). The candidate C2 (gD-μCH3-CH4), showing the most compact oligomer size and uniform distribution of its particles was chosen as the suitable candidate for mice immunization studies to assess the immunogenicity of the antigen gD. The C2 oligomer stimulated a strong anti-gD humoral response with an antibody titer of 102,400 and a strong, biased Th1 immune response in C57BL/6 mice, indicating its potential as a strong immunogen which may serve as an effective vaccine candidate.
Collapse
Affiliation(s)
- Vikas Kumar Singh
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandar Sindri, Ajmer, Rajasthan India 305817
| | - Sandeep Kumar
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandar Sindri, Ajmer, Rajasthan India 305817
| | - Rajeev Kumar Dhaked
- Department of Zoology, Center for Advanced Studies, University of Rajasthan, Jaipur, Rajasthan India 302004
| | - Abdul S. Ansari
- Department of Zoology, Center for Advanced Studies, University of Rajasthan, Jaipur, Rajasthan India 302004
| | - Nirmal K. Lohiya
- Indian Society for the Study of Reproduction and Fertility, Department of Zoology, Center for Advanced Studies, University of Rajasthan, Jaipur, Rajasthan India 302004
| | - Suman Tapryal
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, NH-8, Bandar Sindri, Ajmer, Rajasthan India 305817
| |
Collapse
|
46
|
Kumar Bharathkar S, Parker BW, Malyutin AG, Haloi N, Huey-Tubman KE, Tajkhorshid E, Stadtmueller BM. The structures of secretory and dimeric immunoglobulin A. eLife 2020; 9:56098. [PMID: 33107820 PMCID: PMC7707832 DOI: 10.7554/elife.56098] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Secretory (S) Immunoglobulin (Ig) A is the predominant mucosal antibody, which binds pathogens and commensal microbes. SIgA is a polymeric antibody, typically containing two copies of IgA that assemble with one joining-chain (JC) to form dimeric (d) IgA that is bound by the polymeric Ig-receptor ectodomain, called secretory component (SC). Here, we report the cryo-electron microscopy structures of murine SIgA and dIgA. Structures reveal two IgAs conjoined through four heavy-chain tailpieces and the JC that together form a β-sandwich-like fold. The two IgAs are bent and tilted with respect to each other, forming distinct concave and convex surfaces. In SIgA, SC is bound to one face, asymmetrically contacting both IgAs and JC. The bent and tilted arrangement of complex components limits the possible positions of both sets of antigen-binding fragments (Fabs) and preserves steric accessibility to receptor-binding sites, likely influencing antigen binding and effector functions.
Collapse
Affiliation(s)
- Sonya Kumar Bharathkar
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, United States
| | - Benjamin W Parker
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, United States
| | - Andrey G Malyutin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States.,Beckman Institute, California Institute of Technology, Pasadena, United States
| | - Nandan Haloi
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, United States.,NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Urbana, United States
| | - Kathryn E Huey-Tubman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, United States
| | - Emad Tajkhorshid
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, United States.,Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, United States.,NIH Center for Macromolecular Modeling and Bioinformatics, Beckman Institute for Advanced Science and Technology, Urbana, United States
| | - Beth M Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, United States
| |
Collapse
|
47
|
Jamwal DR, Laubitz D, Harrison CA, da Paz VF, Cox CM, Wong R, Midura-Kiela M, Gurney MA, Besselsen DG, Setty P, Lybarger L, Bhattacharya D, Wilson JM, Ghishan FK, Kiela PR. Intestinal Epithelial Expression of MHCII Determines Severity of Chemical, T-Cell-Induced, and Infectious Colitis in Mice. Gastroenterology 2020; 159:1342-1356.e6. [PMID: 32589883 PMCID: PMC9190026 DOI: 10.1053/j.gastro.2020.06.049] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/14/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Intestinal epithelial cells (IECs) provide a barrier that separates the mucosal immune system from the luminal microbiota. IECs constitutively express low levels of major histocompatibility complex (MHC) class II proteins, which are upregulated upon exposure to interferon gamma. We investigated the effects of deleting MHCII proteins specifically in mice with infectious, dextran sodium sulfate (DSS)-, and T-cell-induced colitis. METHODS We disrupted the histocompatibility 2, class II antigen A, beta 1 gene (H2-Ab1) in IECs of C57BL/6 mice (I-AbΔIEC) or Rag1-/- mice (Rag1-/-I-AbΔIEC); we used I-AbWT mice as controls. Colitis was induced by administration of DSS, transfer of CD4+CD45RBhi T cells, or infection with Citrobacter rodentium. Colon tissues were collected and analyzed by histology, immunofluorescence, xMAP, and reverse-transcription polymerase chain reaction and organoids were generated. Microbiota (total and immunoglobulin [Ig]A-coated) in intestinal samples were analyzed by16S amplicon profiling. IgA+CD138+ plasma cells from Peyer's patches and lamina propria were analyzed by flow cytometry and IgA repertoire was determined by next-generation sequencing. RESULTS Mice with IEC-specific loss of MHCII (I-AbΔIEC mice) developed less severe DSS- or T-cell transfer-induced colitis than control mice. Intestinal tissues from I-AbΔIEC mice had a lower proportion of IgA-coated bacteria compared with control mice, and a reduced luminal concentration of secretory IgA (SIgA) following infection with C rodentium. There was no significant difference in the mucosal IgA repertoire of I-AbΔIEC vs control mice, but opsonization of cultured C rodentium by SIgA isolated from I-AbΔIEC mice was 50% lower than that of SIgA from mAbWT mice. Fifty percent of I-AbΔIEC mice died after infection with C rodentium, compared with none of the control mice. We observed a transient but significant expansion of the pathogen in the feces of I-AbΔIEC mice compared with I-AbWT mice. CONCLUSIONS In mice with DSS or T-cell-induced colitis, loss of MHCII from IECs reduces but does not eliminate mucosal inflammation. However, in mice with C rodentium-induced colitis, loss of MHCII reduces bacterial clearance by decreasing binding of IgA to commensal and pathogenic bacteria.
Collapse
Affiliation(s)
- Deepa R. Jamwal
- Department of Pediatrics, University of Arizona, Tucson, Arizona
| | - Daniel Laubitz
- Department of Pediatrics, University of Arizona, Tucson, Arizona
| | | | | | - Christopher M. Cox
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Rachel Wong
- Department of Immunobiology, University of Arizona, Tucson, Arizona
| | | | | | | | - Prashanth Setty
- Department of Pediatrics, University of Arizona, Tucson, Arizona
| | - Lonnie Lybarger
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | | | - Jean M. Wilson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Fayez K. Ghishan
- Department of Pediatrics, University of Arizona, Tucson, Arizona
| | - Pawel R. Kiela
- Department of Pediatrics, University of Arizona, Tucson, Arizona,Department of Immunobiology, University of Arizona, Tucson, Arizona
| |
Collapse
|
48
|
Sterlin D, Gorochov G. When Therapeutic IgA Antibodies Might Come of Age. Pharmacology 2020; 106:9-19. [PMID: 32950975 DOI: 10.1159/000510251] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/10/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Extensive efforts have been made in optimizing monoclonal immunoglobulin (Ig)G antibodies for use in clinical practice. Accumulating evidence suggests that IgA or anti-FcαRI could also represent an exciting avenue toward novel therapeutic strategies. SUMMARY Here, we underline that IgA is more effective in recruiting neutrophils for tumor cell killing and is potently active against several pathogens, including rotavirus, poliovirus, influenza virus, and SARS-CoV-2. IgA could also be used to modulate excessive immune responses in inflammatory diseases. Furthermore, secretory IgA is emerging as a major regulator of gut microbiota, which impacts intestinal homeostasis and global health as well. As such, IgA could be used to promote a healthy microbiota in a therapeutic setting. Key messages: IgA combines multifaceted functions that can be desirable for immunotherapy.
Collapse
Affiliation(s)
- Delphine Sterlin
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, AP-HP Hôpital Pitié-Salpêtrière, Paris, France.,Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR1222 Inserm, Paris, France
| | - Guy Gorochov
- Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Sorbonne Université, Inserm, AP-HP Hôpital Pitié-Salpêtrière, Paris, France,
| |
Collapse
|
49
|
Murin CD. Considerations of Antibody Geometric Constraints on NK Cell Antibody Dependent Cellular Cytotoxicity. Front Immunol 2020; 11:1635. [PMID: 32849559 PMCID: PMC7406664 DOI: 10.3389/fimmu.2020.01635] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/18/2020] [Indexed: 12/31/2022] Open
Abstract
It has been well-established that antibody isotype, glycosylation, and epitope all play roles in the process of antibody dependent cellular cytotoxicity (ADCC). For natural killer (NK) cells, these phenotypes are linked to cellular activation through interaction with the IgG receptor FcγRIIIa, a single pass transmembrane receptor that participates in cytoplasmic signaling complexes. Therefore, it has been hypothesized that there may be underlying spatial and geometric principles that guide proper assembly of an activation complex within the NK cell immune synapse. Further, synergy of antibody phenotypic properties as well as allosteric changes upon antigen binding may also play an as-of-yet unknown role in ADCC. Understanding these facets, however, remains hampered by difficulties associated with studying immune synapse dynamics using classical approaches. In this review, I will discuss relevant NK cell biology related to ADCC, including the structural biology of Fc gamma receptors, and how the dynamics of the NK cell immune synapse are being studied using innovative microscopy techniques. I will provide examples from the literature demonstrating the effects of spatial and geometric constraints on the T cell receptor complex and how this relates to intracellular signaling and the molecular nature of lymphocyte activation complexes, including those of NK cells. Finally, I will examine how the integration of high-throughput and "omics" technologies will influence basic NK cell biology research moving forward. Overall, the goal of this review is to lay a basis for understanding the development of drugs and therapeutic antibodies aimed at augmenting appropriate NK cell ADCC activity in patients being treated for a wide range of illnesses.
Collapse
Affiliation(s)
- Charles D. Murin
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, United States
| |
Collapse
|
50
|
Perruzza L, Jaconi S, Lombardo G, Pinna D, Strati F, Morone D, Seehusen F, Hu Y, Bajoria S, Xiong J, Kumru OS, Joshi SB, Volkin DB, Piantanida R, Benigni F, Grassi F, Corti D, Pizzuto MS. Prophylactic Activity of Orally Administered FliD-Reactive Monoclonal SIgA Against Campylobacter Infection. Front Immunol 2020; 11:1011. [PMID: 32582158 PMCID: PMC7296071 DOI: 10.3389/fimmu.2020.01011] [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] [Received: 02/27/2020] [Accepted: 04/28/2020] [Indexed: 12/17/2022] Open
Abstract
Campylobacter infection is one of the most common causes of bacterial gastroenteritis worldwide and a major global health threat due to the rapid development of antibiotic resistance. Currently, there are no vaccines approved to prevent campylobacteriosis, and rehydration is the main form of therapy. Secretory immunoglobulin A (SIgA) is the main antibody class found in mucous secretions, including human milk, and serves as the first line of defense for the gastrointestinal epithelium against enteric pathogens. In this study, we describe the prophylactic activity of orally delivered recombinant SIgA generated from two human monoclonal antibodies (CAA1 and CCG4) isolated for their reactivity against the flagellar-capping protein FliD, which is essential for bacteria motility and highly conserved across Campylobacter species associated with severe enteritis. In an immunocompetent weaned mouse model, a single oral administration of FliD-reactive SIgA CAA1 or CCG4 at 2 h before infection significantly enhances Campylobacter clearance at early stages post-infection, reducing the levels of inflammation markers associated with epithelial damage and polymorphonuclear (PMN) cells infiltration in the cecum lamina propria. Our data indicate that the prophylactic activity of CAA1 and CCG4 is not only dependent on the specificity to FliD but also on the use of the SIgA format, as the immunoglobulin G (IgG) versions of the same antibodies did not confer a comparable protective effect. Our work emphasizes the potential of FliD as a target for the development of vaccines and supports the concept that orally administered FliD-reactive SIgA can be developed to prevent or mitigate the severity of Campylobacter infections as well as the development of post-infection syndromes.
Collapse
Affiliation(s)
- Lisa Perruzza
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Stefano Jaconi
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Gloria Lombardo
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Debora Pinna
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Francesco Strati
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Diego Morone
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Frauke Seehusen
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Yue Hu
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Jian Xiong
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Ozan Selahattin Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Sangeeta Bagai Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - David Bernard Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, United States
| | - Renato Piantanida
- Department of Otolaryngology-Head and Neck Surgery, Ospedale Regionale di Lugano, Lugano, Switzerland
| | - Fabio Benigni
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | - Fabio Grassi
- Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Università della Svizzera Italiana (USI), Bellinzona, Switzerland
| | - Davide Corti
- Humabs BioMed SA a Subsidiary of Vir Biotechnology Inc., Bellinzona, Switzerland
| | | |
Collapse
|