1
|
Abad P, Marín-García P, Heras M, Fobil JN, Hutchful AG, Diez A, Puyet A, Reyes-Palomares A, Azcárate IG, Bautista JM. Microscopic and submicroscopic infection by Plasmodium falciparum: Immunoglobulin M and A profiles as markers of intensity and exposure. Front Cell Infect Microbiol 2022; 12:934321. [PMID: 36118030 PMCID: PMC9478039 DOI: 10.3389/fcimb.2022.934321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 08/04/2022] [Indexed: 11/15/2022] Open
Abstract
Assessment of serological Plasmodium falciparum–specific antibodies in highly endemic areas provides valuable information about malaria status and parasite exposure in the population. Although serological evidence of Plasmodium exposure is commonly determined by Plasmodium-specific immunoglobulin G (IgG) levels; IgM and IgA are likely markers of malaria status that remain relatively unexplored. Previous studies on IgM and IgA responses have been based on their affinity for single antigens with shortage of immune responses analysis against the whole Plasmodium proteome. Here, we provide evidence of how P. falciparum infection triggers the production of specific IgM and IgA in plasma and its relationship with parasite density and changes in hematological parameters. A total of 201 individuals attending a hospital in Breman Asikuma, Ghana, were recruited into this study. Total and P. falciparum–specific IgM, IgA, and IgG were assessed by ELISA and examined in relation to age (0–5, 14–49, and ≥50 age ranges); infection (submicroscopic vs. microscopic malaria); pregnancy and hematological parameters. Well-known IgG response was used as baseline control. P. falciparum–specific IgM and IgA levels increased in the population with the age, similarly to IgG. These data confirm that acquired humoral immunity develops by repeated infections through the years endorsing IgM and IgA as exposure markers in endemic malaria regions. High levels of specific IgA and IgM in children were associated with microscopic malaria and worse prognosis, because most of them showed severe anemia. This new finding shows that IgM and IgA may be used as diagnostic markers in this age group. We also found an extremely high prevalence of submicroscopic malaria (46.27% on average) accompanied by IgM and IgA levels indistinguishable from those of uninfected individuals. These data, together with the observed lack of sensitivity of rapid diagnostic tests (RDTs) compared to PCR, invoke the urgent need to implement diagnostic markers for submicroscopic malaria. Overall, this study opens the potential use of P. falciparum–specific IgM and IgA as new serological markers to predict malaria status in children and parasite exposure in endemic populations. The difficulties in finding markers of submicroscopic malaria are highlighted, emphasizing the need to explore this field in depth.
Collapse
Affiliation(s)
- Paloma Abad
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | | | - Marcos Heras
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Julius N. Fobil
- Department of Biological, Environmental and Occupational Health Sciences, School of Public Health, College of Health Sciences, University of Ghana, Legon, Accra, Ghana
| | - Alfred G. Hutchful
- Laboratory of Hematology and Infectious Diseases, Our Lady of Grace Hospital, Breman-Asikuma, Ghana
| | - Amalia Diez
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Antonio Puyet
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Armando Reyes-Palomares
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
| | - Isabel G. Azcárate
- Faculty of Health Sciences, Rey Juan Carlos University, Alcorcón, Spain
- *Correspondence: Isabel G. Azcárate, ; José M. Bautista,
| | - José M. Bautista
- Department of Biochemistry and Molecular Biology and Research Institute Hospital 12 de Octubre (Imas12), Universidad Complutense de Madrid, Madrid, Spain
- *Correspondence: Isabel G. Azcárate, ; José M. Bautista,
| |
Collapse
|
2
|
Aljabban J, Rohr M, Syed S, Khorfan K, Borkowski V, Aljabban H, Segal M, Mukhtar M, Mohammed M, Panahiazar M, Hadley D, Spengler R, Spengler E. Transcriptome changes in stages of non-alcoholic fatty liver disease. World J Hepatol 2022; 14:1382-1397. [PMID: 36158924 PMCID: PMC9376779 DOI: 10.4254/wjh.v14.i7.1382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/29/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the United States and globally. The currently understood model of pathogenesis consists of a ‘multiple hit’ hypothesis in which environmental and genetic factors contribute to hepatic inflammation and injury.
AIM To examine the genetic expression of NAFLD and non-alcoholic steatohepatitis (NASH) tissue samples to identify common pathways that contribute to NAFLD and NASH pathogenesis.
METHODS We employed the Search Tag Analyze Resource for Gene Expression Omnibus platform to search the The National Center for Biotechnology Information Gene Expression Omnibus to elucidate NAFLD and NASH pathology. For NAFLD, we conducted meta-analysis of data from 58 NAFLD liver biopsies and 60 healthy liver biopsies; for NASH, we analyzed 187 NASH liver biopsies and 154 healthy liver biopsies.
RESULTS Our results from the NAFLD analysis reinforce the role of altered metabolism, inflammation, and cell survival in pathogenesis and support recently described contributors to disease activity, such as altered androgen and long non-coding RNA activity. The top upstream regulator was found to be sterol regulatory element binding transcription factor 1 (SREBF1), a transcription factor involved in lipid homeostasis. Downstream of SREBF1, we observed upregulation in CXCL10, HMGCR, HMGCS1, fatty acid binding protein 5, paternally expressed imprinted gene 10, and downregulation of sex hormone-binding globulin and insulin-like growth factor 1. These molecular changes reflect low-grade inflammation secondary to accumulation of fatty acids in the liver. Our results from the NASH analysis emphasized the role of cholesterol in pathogenesis. Top canonical pathways, disease networks, and disease functions were related to cholesterol synthesis, lipid metabolism, adipogenesis, and metabolic disease. Top upstream regulators included pro-inflammatory cytokines tumor necrosis factor and IL1B, PDGF BB, and beta-estradiol. Inhibition of beta-estradiol was shown to be related to derangement of several cellular downstream processes including metabolism, extracellular matrix deposition, and tumor suppression. Lastly, we found riciribine (an AKT inhibitor) and ZSTK-474 (a PI3K inhibitor) as potential drugs that targeted the differential gene expression in our dataset.
CONCLUSION In this study we describe several molecular processes that may correlate with NAFLD disease and progression. We also identified ricirbine and ZSTK-474 as potential therapy.
Collapse
Affiliation(s)
- Jihad Aljabban
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Michael Rohr
- Department of Medicine, University of Central Florida College of Medicine, Orlando, FL 32827, United States
| | - Saad Syed
- Department of Medicine, Northwestern Memorial Hospital, Chicago, IL 60611, United States
| | - Kamal Khorfan
- Department of Gastroenterology and Hepatology, University of California San Francisco-Fresno , Fresno, CA 93701, United States
| | - Vincent Borkowski
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Hisham Aljabban
- Department of Medicine, Barry University, Miami, FL 33161, United States
| | - Michael Segal
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Mohamed Mukhtar
- Department of Medicine, Michigan State University College of Human Medicine, East Lansing, MI 49503, United States
| | - Mohammed Mohammed
- Department of Medicine, Windsor University School of Medicine, Saint Kitts 1621, Cayon, Saint Kitts and Nevis
| | - Maryam Panahiazar
- Department of Surgery, University of California San Francisco, San Francisco, CA 94305, United States
| | - Dexter Hadley
- Department of Artificial Intelligence, Pathology, University of Central Florida College of Medicine , Orlando, FL 32827, United States
| | - Ryan Spengler
- Department of Medicine, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| | - Erin Spengler
- Department of Gastroenterology and Hepatology, University of Wisconsin Hospital and Clinics, Madison, WI 53792, United States
| |
Collapse
|
3
|
Li N, Wang J, Zhan X. Identification of Immune-Related Gene Signatures in Lung Adenocarcinoma and Lung Squamous Cell Carcinoma. Front Immunol 2021; 12:752643. [PMID: 34887858 PMCID: PMC8649721 DOI: 10.3389/fimmu.2021.752643] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/22/2021] [Indexed: 12/25/2022] Open
Abstract
Accumulating evidence indicates that immunotherapy helped to improve the survival and quality-of-life of patients with lung adenocarcinoma (LUAD) or lung squamous cell carcinoma (LUSC) besides chemotherapy and gene targeting treatment. This study aimed to develop immune-related gene signatures in LUAD and LUSC subtypes, respectively. LUAD and LUSC samples were divided into high- and low-abundance groups of immune cell infiltration (Immunity_H and Immunity_L) based on the abundance of immune cell infiltrations. The distribution of immune cells was significantly different between the high- and low-immunity subtypes in LUAD and LUSC samples. The differentially expressed genes (DEGs) between those two groups in LUAD and LUSC contain some key immune-related genes, such as PDL1, PD1, CTLA-4, and HLA families. The DEGs were enriched in multiple immune-related pathways. Furthermore, the seven-immune-related-gene-signature (CD1B, CHRNA6, CLEC12B, CLEC17A, CLNK, INHA, and SLC14A2) prognostic model-based high- and low-risk groups were significantly associated with LUAD overall survival and clinical characteristics. The eight-immune-related-gene-signature (C4BPB, FCAMR, GRAPL, MAP1LC3C, MGC2889, TRIM55, UGT1A1, and VIPR2) prognostic model-based high- and low-risk groups were significantly associated with LUSC overall survival and clinical characteristics. The prognostic models were tested as good ones by receiver operating characteristic, principal component analysis, univariate and multivariate analysis, and nomogram. The verifications of these two immune-related-gene-signature prognostic models showed consistency in the train and test cohorts of LUAD and LUSC. In addition, patients with LUAD in the low-risk group responded better to immunotherapy than those in the high-risk group. This study revealed two reliable immune-related-gene-signature models that were significantly associated with prognosis and tumor microenvironment cell infiltration in LUAD and LUSC, respectively. Evaluation of the integrated characterization of multiple immune-related genes and pathways could help to predict the response to immunotherapy and monitor immunotherapy strategies.
Collapse
Affiliation(s)
- Na Li
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan, China.,Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China
| | - Jiahong Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xianquan Zhan
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan, China.,Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, China.,Gastroenterology Research Institute and Clinical Center, Shandong First Medical University, Jinan, China
| |
Collapse
|
4
|
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: 5] [Impact Index Per Article: 1.3] [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
|
5
|
Mourcin F, Verdière L, Roulois D, Amin R, Lamaison C, Sibut V, Thamphya B, Pangault C, Monvoisin C, Huet S, Seffals M, Baulande S, Mechta-Grigoriou F, Legoix P, Rossille D, Guirriec M, Léonard S, Cartron G, Salles G, Fest T, Tarte K. Follicular lymphoma triggers phenotypic and functional remodeling of the human lymphoid stromal cell landscape. Immunity 2021; 54:1788-1806.e7. [DOI: 10.1016/j.immuni.2021.05.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/18/2021] [Accepted: 05/27/2021] [Indexed: 02/08/2023]
|
6
|
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
|
7
|
Chen K, Magri G, Grasset EK, Cerutti A. Rethinking mucosal antibody responses: IgM, IgG and IgD join IgA. Nat Rev Immunol 2020; 20:427-441. [PMID: 32015473 PMCID: PMC10262260 DOI: 10.1038/s41577-019-0261-1] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2019] [Indexed: 02/08/2023]
Abstract
Humoral immune responses at mucosal surfaces have historically focused on IgA. Growing evidence highlights the complexity of IgA-inducing pathways and the functional impact of IgA on mucosal commensal bacteria. In the gut, IgA contributes to the establishment of a mutualistic host-microbiota relationship that is required to maintain homeostasis and prevent disease. This Review discusses how mucosal IgA responses occur in an increasingly complex humoral defence network that also encompasses IgM, IgG and IgD. Aside from integrating the protective functions of IgA, these hitherto neglected mucosal antibodies may strengthen the communication between mucosal and systemic immune compartments.
Collapse
Affiliation(s)
- Kang Chen
- Department of Obstetrics and Gynecology and Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Giuliana Magri
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona Biomedical Research Park, Barcelona, Spain
| | - Emilie K Grasset
- The Immunology Institute, Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Cerutti
- Institut Hospital del Mar d'Investigacions Mèdiques (IMIM), Barcelona Biomedical Research Park, Barcelona, Spain.
- The Immunology Institute, Department of Medicine, Mount Sinai School of Medicine, New York, NY, USA.
- Catalan Institute for Research and Advanced Studies (ICREA), Barcelona Biomedical Research Park, Barcelona, Spain.
| |
Collapse
|
8
|
Lyashchenko KP, Vordermeier HM, Waters WR. Memory B cells and tuberculosis. Vet Immunol Immunopathol 2020; 221:110016. [PMID: 32050091 DOI: 10.1016/j.vetimm.2020.110016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/30/2019] [Accepted: 01/29/2020] [Indexed: 02/09/2023]
Abstract
Immunological memory is a central feature of adaptive immunity. Memory B cells are generated upon stimulation with antigen presented by follicular dendritic cells in the peripheral lymphoid tissues. This process typically involves class-switch recombination and somatic hypermutation and it can be dependent or independent on germinal centers or T cell help. The mature B cell memory pool is generally characterized by remarkable heterogeneity of functionally and phenotypically distinct sub-populations supporting multi-layer immune plasticity. Memory B cells found in human patients infected with Mycobacterium tuberculosis include IgD+ CD27+ and IgM+ CD27+ subsets. In addition, expansion of atypical memory B cells characterized by the lack of CD27 expression and by inability to respond to antigen-induced re-activation is documented in human tuberculosis. These functionally impaired memory B cells are believed to have adverse effects on host immunity. Human and animal studies demonstrate recruitment of antigen-activated B cells to the infection sites and their presence in lung granulomas where proliferating B cells are organized into discrete clusters resembling germinal centers of secondary lymphoid organs. Cattle studies show development of IgM+, IgG+, and IgA+ memory B cells in M. bovis infection with the ability to rapidly differentiate into antibody-producing plasma cells upon antigen re-exposure. This review discusses recent advances in research on generation, re-activation, heterogeneity, and immunobiological functions of memory B cells in tuberculosis. The role of memory B cells in post-skin test recall antibody responses in bovine tuberculosis and implications for development of improved immunodiagnostics are also reviewed.
Collapse
Affiliation(s)
| | - H Martin Vordermeier
- Tuberculosis Research Group, Animal and Plant Health Agency, Addlestone, United Kingdom; Institute for Biological, Environmental and Rural Sciences, Aberystwyth University, Aberystwyth, United Kingdom
| | - W Ray Waters
- National Animal Disease Center, Agricultural Research Service, US Department of Agriculture, Ames, IA, USA
| |
Collapse
|
9
|
Michaud E, Mastrandrea C, Rochereau N, Paul S. Human Secretory IgM: An Elusive Player in Mucosal Immunity. Trends Immunol 2020; 41:141-156. [PMID: 31928913 DOI: 10.1016/j.it.2019.12.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 12/29/2022]
Abstract
Secretory IgMs (SIgMs) were amongst the first identified immunoglobulins. However, their importance was not fully understood and recent advances have shown they play a key role in establishing and promoting commensal gut tolerance in mice and humans. The true interactions between SIgMs and the microbiota remain controversial and we aim to consolidate current knowledge in this review. Through comprehensive examination of SIgMs and their corresponding B cell secretors in several different pathological immunological contexts, we review the presumed role of these molecules in gut tolerance, inflammatory bowel diseases, and lung immunity. As SIgMs harbor a mostly tolerogenic function, we posit that their inclusion in further immunological research is paramount.
Collapse
Affiliation(s)
- Eva Michaud
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | | | - Nicolas Rochereau
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Stéphane Paul
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France.
| |
Collapse
|
10
|
Regulation of Humoral Immune Responses and B Cell Tolerance by the IgM Fc Receptor (FcμR). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1254:75-86. [DOI: 10.1007/978-981-15-3532-1_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
11
|
Goulet DR, Atkins WM. Considerations for the Design of Antibody-Based Therapeutics. J Pharm Sci 2020; 109:74-103. [PMID: 31173761 PMCID: PMC6891151 DOI: 10.1016/j.xphs.2019.05.031] [Citation(s) in RCA: 173] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/02/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Antibody-based proteins have become an important class of biologic therapeutics, due in large part to the stability, specificity, and adaptability of the antibody framework. Indeed, antibodies not only have the inherent ability to bind both antigens and endogenous immune receptors but also have proven extremely amenable to protein engineering. Thus, several derivatives of the monoclonal antibody format, including bispecific antibodies, antibody-drug conjugates, and antibody fragments, have demonstrated efficacy for treating human disease, particularly in the fields of immunology and oncology. Reviewed here are considerations for the design of antibody-based therapeutics, including immunological context, therapeutic mechanisms, and engineering strategies. First, characteristics of antibodies are introduced, with emphasis on structural domains, functionally important receptors, isotypic and allotypic differences, and modifications such as glycosylation. Then, aspects of therapeutic antibody design are discussed, including identification of antigen-specific variable regions, choice of expression system, use of multispecific formats, and design of antibody derivatives based on fragmentation, oligomerization, or conjugation to other functional moieties. Finally, strategies to enhance antibody function through protein engineering are reviewed while highlighting the impact of fundamental biophysical properties on protein developability.
Collapse
Affiliation(s)
- Dennis R Goulet
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195.
| | - William M Atkins
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195
| |
Collapse
|
12
|
Liu J, Wang Y, Xiong E, Hong R, Lu Q, Ohno H, Wang JY. Role of the IgM Fc Receptor in Immunity and Tolerance. Front Immunol 2019; 10:529. [PMID: 30967868 PMCID: PMC6438924 DOI: 10.3389/fimmu.2019.00529] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 02/27/2019] [Indexed: 11/13/2022] Open
Abstract
Immunoglobulin (Ig) M is the first antibody isotype to appear during evolution, ontogeny and immune responses. IgM not only serves as the first line of host defense against infections but also plays an important role in immune regulation and immunological tolerance. For many years, IgM is thought to function by binding to antigen and activating complement system. With the discovery of the IgM Fc receptor (FcμR), it is now clear that IgM can also elicit its function through FcμR. In this review, we will describe the molecular characteristics of FcμR, its role in B cell development, maturation and activation, humoral immune responses, host defense, and immunological tolerance. We will also discuss the functional relationship between IgM-complement and IgM-FcμR pathways in regulating immunity and tolerance. Finally, we will discuss the potential involvement of FcμR in human diseases.
Collapse
Affiliation(s)
- Jun Liu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ermeng Xiong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Rongjian Hong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hiroshi Ohno
- RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Tang CH, Chang S, Hashimoto A, Chen YJ, Kang CW, Mato AR, Del Valle JR, Gabrilovich DI, Hu CC. Secretory IgM Exacerbates Tumor Progression by Inducing Accumulations of MDSCs in Mice. Cancer Immunol Res 2018; 6:696-710. [PMID: 29650518 DOI: 10.1158/2326-6066.cir-17-0582] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/06/2018] [Accepted: 04/05/2018] [Indexed: 12/15/2022]
Abstract
Chronic lymphocytic leukemia (CLL) cells can secrete immunoglobulin M. However, it is not clear whether secretory IgM (sIgM) plays a role in disease progression. We crossed the Eμ-TCL1 mouse model of CLL, in which the expression of human TCL1 oncogene was driven by the V(H) promoter-Ig(H)-Eμ enhancer, with MD4 mice whose B cells produced B-cell receptor (membrane-bound IgM) and sIgM with specificity for hen egg lysozyme (HEL). CLL cells that developed in these MD4/Eμ-TCL1 mice reactivated a parental Ig gene allele and secreted IgM, and did not recognize HEL. The MD4/Eμ-TCL1 mice had reduced survival, increased myeloid-derived suppressor cells (MDSC), and decreased numbers of T cells. We tested whether sIgM could contribute to the accumulation of MDSCs by crossing μS-/- mice, which could not produce sIgM, with Eμ-TCL1 mice. The μS-/-/Eμ-TCL1 mice survived longer than Eμ-TCL1 mice and developed decreased numbers of MDSCs which were less able to suppress proliferation of T cells. We targeted the synthesis of sIgM by deleting the function of XBP-1s and showed that targeting XBP-1s genetically or pharmacologically could lead to decreased sIgM, accompanied by decreased numbers and reduced functions of MDSCs in MD4/Eμ-TCL1 mice. Additionally, MDSCs from μS-/- mice grafted with Lewis lung carcinoma were inefficient suppressors of T cells, resulting in slower tumor growth. These results demonstrate that sIgM produced by B cells can upregulate the functions of MDSCs in tumor-bearing mice to aggravate cancer progression. In a mouse model of CLL, production of secretory IgM led to more MDSCs, fewer T cells, and shorter survival times for the mice. Thus, secretory IgM may aggravate the progression of this cancer. Cancer Immunol Res; 6(6); 696-710. ©2018 AACR.
Collapse
Affiliation(s)
| | - Shiun Chang
- The Wistar Institute, Philadelphia, Pennsylvania
| | | | - Yi-Ju Chen
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Chang Won Kang
- Department of Chemistry, University of South Florida, Tampa, Florida
| | - Anthony R Mato
- Center for CLL, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Juan R Del Valle
- Department of Chemistry, University of South Florida, Tampa, Florida
| | | | - Chih-Chi Hu
- The Wistar Institute, Philadelphia, Pennsylvania.
| |
Collapse
|
14
|
Blackburn TE, Santiago T, Burrows PD. FCRLA-A Resident Endoplasmic Reticulum Protein that Associates with Multiple Immunoglobulin Isotypes in B Lineage Cells. Curr Top Microbiol Immunol 2017; 408:47-65. [PMID: 28879521 DOI: 10.1007/82_2017_40] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
FCRLA is homologous to receptors for the Fc portion of IgG (FcγR) and is located in the same region of human chromosome one, but has several unusual and unique features. It is a soluble resident ER protein retained in this organelle by unknown mechanisms involving the N-terminal domain, a disordered domain with three Cys residues in close proximity in the human protein. Unlike the FcγRs, FCRLA is not glycosylated and has no transmembrane region. FCRLA is included in this CTMI volume on IgM-binding proteins because it binds IgM in the ER, but quite surprisingly, given the isotype-restricted ligand specificity of the other FcRs, it also binds all other Ig isotypes so far tested, IgG and IgA. In the case of IgM, there is even preferential binding of the secretory and not the transmembrane form. Among B cells, FCRLA is most highly expressed in the germinal center and shows little expression in plasma cells. Based on these observations, we propose that one human FCRLA function is to stop GC B cells from secreting IgM, which would act as a decoy receptor, thus preventing the B cells from capturing antigen, processing it, and presenting the antigen-derived peptides to T follicular helper cells. Without help from these T cells, there would be limited B cell isotype switching, proliferation, and differentiation. On the other hand, FCRLA is downregulated in plasma cells, where IgM secretion is an essential function. FCRLA may also act as a chaperone involved by unknown mechanisms in the proper assembly of Ig molecules of all isotypes.
Collapse
Affiliation(s)
- Tessa E Blackburn
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Teresa Santiago
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter D Burrows
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
15
|
Affiliation(s)
- P Mark Hogarth
- Centre for Biomedicine, Burnet Institute, Melbourne, Vic., Australia.,Department of Immunology, Monash University, Melbourne, Vic., Australia.,Department of Pathology, University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|