1
|
Hirayama M, Ohyama Y, Tsuji Y, Enomoto T, Hasegawa M, Tsuboi N, Novak J, Takahashi K. Longitudinal changes in the abundance of IgA1 O- and N-glycoforms in IgA nephropathy. Clin Exp Nephrol 2025:10.1007/s10157-025-02659-y. [PMID: 40195177 DOI: 10.1007/s10157-025-02659-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND IgA nephropathy (IgAN) is the most common type of primary glomerulonephritis. Elevation in the blood levels of aberrantly glycosylated IgA1 is a crucial initial step in IgAN pathogenesis. Here, we aimed to determine the longitudinal changes in the serum levels of IgA1 O- and N-glycoforms in patients with IgAN receiving different treatments. METHODS We enrolled Japanese patients diagnosed with primary IgAN: 10 patients who underwent tonsillectomy and corticosteroid therapy (T-CST), 7 who received corticosteroid therapy (CST), 8 who received conservative therapy (CO), and 5 with other renal diseases who received corticosteroid therapy (ORD) as disease controls. IgA was purified from patient sera collected at diagnosis and post-treatment. After sample preparation, O-glycoforms of the hinge region (HR) and N-glycoforms of the fragment crystallizable region were analyzed using high-resolution mass spectrometry (MS). RESULTS The MS analysis of O-glycoforms of IgA1 showed that the relative abundance of IgA1 with 3GalNAc3Gal, which we previously identified as a characteristic IgA1 O-glycoform in IgAN, decreased post-treatment only in the T-CST group (P = 0.0195). Regarding N-glycoforms, the relative abundance of fucosylated N-glycan at asparagine (Asn)340 increased in the IgAN group compared with that in the ORD group (P = 0.0189) and decreased post-treatment only in the T-CST group (P = 0.0195). CONCLUSION The MS analysis of O- and N-glycoforms of IgA1 revealed substantial changes in their abundance in the T-CST group but not in the CST, CO, and ORD groups. Our study provides new insights into how specific treatments alter the IgA1 glycoform abundance.
Collapse
Affiliation(s)
- Masaya Hirayama
- Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
- Department of Pathology and Cytopathology, Fujita Health University School of Medical Sciences, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Yukako Ohyama
- Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Yudai Tsuji
- Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Tetsuro Enomoto
- Oriental Yeast Co., Ltd, 50 Kanou-Cho, Nagahama, Shiga, 526-0804, Japan
| | - Midori Hasegawa
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Naotake Tsuboi
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL, 35294, USA
| | - Kazuo Takahashi
- Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan.
- Department of Nephrology, Fujita Health University School of Medicine, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
2
|
Shillitoe B, Duque JSR, Lai SHY, Lau TM, Chan JCH, Bourne H, Stroud C, Flood T, Buckland M, Ip W, Worth A, Hackett S, Herwadkar A, Coulter T, Blaney C, Jolles S, Garcez T, Moya E, Faust S, Pearce MS, Lau YL, Gennery AR. Outcomes of X-Linked Agammaglobulinaemia Patients. J Clin Immunol 2024; 45:40. [PMID: 39541002 DOI: 10.1007/s10875-024-01829-z] [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: 04/15/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND X-linked agammaglobulinaemia (XLA), caused by mutations in BTK, is characterised by low or absent peripheral CD19 + B lymphocytes and agammaglobulinaemia. The mainstay of treatment consists of immunoglobulin replacement therapy (IgRT). As this cannot fully compensate for the immune defects in XLA, patients may therefore continue to be at risk of complications. OBJECTIVES To describe the clinical outcomes of XLA patients in the United Kingdom and Hong Kong and evaluate current treatment strategies. METHODS Patients with a definitive diagnosis of XLA were included in this cross-sectional and retrospective analysis of clinical health outcomes. Data pertaining to diagnosis, infection incidence, IgG trough levels and lung function were collected and analysed. RESULTS 99 patients with a median age of 29.02 years (IQR 12.83-37.41) and a total follow up of 1922 patient years, were included this study. The median age at diagnosis was 3.30 years (IQR 1.04-8.38) which decreased over time (p = 0.004). 40% of the cohort had radiological evidence of bronchiectasis. Risk of bronchiectasis was not significantly associated with clinical infection incidence (p = 0.880) or IgG trough levels (p = 0.407). Two patients demonstrated novel complications, namely persistent norovirus infection, leading to haemopoietic stem cell transplantation (HSCT). CONCLUSIONS Despite modern therapy, most XLA patients continue to experience complications, most notably bronchiectasis, likely due to absence of IgA/M in current therapies, but lack of B lymphocytes may also lead to additional sequalae. These data strongly support the need for further research, particularly that of curative modalities including HSCT and gene therapy.
Collapse
Affiliation(s)
- Ben Shillitoe
- Sheffield Children's NHS Foundation Trust, Sheffield, UK.
| | - Jaime S Rosa Duque
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Sophie H Y Lai
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Tsun Ming Lau
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Jeffery C H Chan
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Helen Bourne
- Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Catherine Stroud
- Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Terry Flood
- Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Matthew Buckland
- Institute of Child Health, UCL, London, UK
- University College London, London, UK
| | - Winnie Ip
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Austen Worth
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Scott Hackett
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Archana Herwadkar
- Salford Care Organisation, Northern Care Alliance NHS Trust, Manchester, UK
| | | | | | - Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK
| | - Tomaz Garcez
- Manchester University NHS Foundation Trust, Manchester, UK
| | - Eduardo Moya
- Bradford Teaching Hospitals NHS Foundation Trust, Bradford, UK
| | - Saul Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Faculty of Medicine, Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Mark S Pearce
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China.
| | - Andrew R Gennery
- Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
3
|
Uchino T, Uchida M, Ito R, Fujie S, Iemitsu K, Kojima C, Nakamura M, Shimizu K, Tanimura Y, Shinohara Y, Hashimoto T, Isaka T, Iemitsu M. Effects of different exercise intensities or durations on salivary IgA secretion. Eur J Appl Physiol 2024; 124:2687-2696. [PMID: 38634902 PMCID: PMC11365859 DOI: 10.1007/s00421-024-05467-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 03/09/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE This study aimed to examine changes in salivary immunoglobulin A (s-IgA) secretion at different intensities or durations of acute exercise. METHODS Twelve healthy untrained young males were included in randomized crossover trials in Experiment 1 (cycling exercise for 30 min at a work rate equivalent to 35%, 55%, and 75% maximal oxygen uptake [ V ˙ O2max]) and Experiment 2 (cycling exercise at 55% V ˙ O2max intensity for 30, 60, and 90 min). Saliva samples were collected at baseline, immediately after, and 60 min after each exercise. RESULTS Experiment 1: The percentage change in the s-IgA secretion rate in the 75% V ˙ O2max trial was significantly lower than that in the 55% V ˙ O2max trial immediately after exercise (- 45.7%). The percentage change in the salivary concentration of cortisol, an s-IgA regulating factor, immediately after exercise significantly increased compared to that at baseline in the 75% V ˙ O2max trial (+ 107.6%). A significant negative correlation was observed between the percentage changes in saliva flow rate and salivary cortisol concentration (r = - 0.52, P < 0.01). Experiment 2: The percentage change in the s-IgA secretion rate in the 90-min trial was significantly lower than that in the 30-min trial immediately after exercise (-37.0%). However, the percentage change in salivary cortisol concentration remained the same. CONCLUSION Our findings suggest that a reduction in s-IgA secretion is induced by exercise intensity of greater than or equal to 75% V ˙ O2max for 30 min or exercise duration of greater than or equal to 90 min at 55% V ˙ O2max in healthy untrained young men.
Collapse
Affiliation(s)
- Takamasa Uchino
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Masataka Uchida
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Shiga, Japan
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Reita Ito
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Shumpei Fujie
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Keiko Iemitsu
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Chihiro Kojima
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Shiga, Japan
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Mariko Nakamura
- Department of Sport Science and Research, Japan Institute of Sports Sciences, Nishigaoka Kita-ku, Tokyo, Japan
| | - Kazuhiro Shimizu
- Department of Sport Science and Research, Japan Institute of Sports Sciences, Nishigaoka Kita-ku, Tokyo, Japan
| | - Yuko Tanimura
- Department of Sport Science and Research, Japan Institute of Sports Sciences, Nishigaoka Kita-ku, Tokyo, Japan
| | - Yasushi Shinohara
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Takeshi Hashimoto
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Tadao Isaka
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Motoyuki Iemitsu
- Faculty of Sport and Health Science, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
- Institute of Advanced Research for Sport and Health Science, Ritsumeikan University, Kusatsu, Shiga, Japan.
| |
Collapse
|
4
|
Kazer SW, Match CM, Langan EM, Messou MA, LaSalle TJ, O'Leary E, Marbourg J, Naughton K, von Andrian UH, Ordovas-Montanes J. Primary nasal influenza infection rewires tissue-scale memory response dynamics. Immunity 2024; 57:1955-1974.e8. [PMID: 38964332 PMCID: PMC11324402 DOI: 10.1016/j.immuni.2024.06.005] [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: 05/05/2023] [Revised: 03/14/2024] [Accepted: 06/10/2024] [Indexed: 07/06/2024]
Abstract
The nasal mucosa is often the initial site of respiratory viral infection, replication, and transmission. Understanding how infection shapes tissue-scale primary and memory responses is critical for designing mucosal therapeutics and vaccines. We generated a single-cell RNA-sequencing atlas of the murine nasal mucosa, sampling three regions during primary influenza infection and rechallenge. Compositional analysis revealed restricted infection to the respiratory mucosa with stepwise changes in immune and epithelial cell subsets and states. We identified and characterized a rare subset of Krt13+ nasal immune-interacting floor epithelial (KNIIFE) cells, which concurrently increased with tissue-resident memory T (TRM)-like cells. Proportionality analysis, cell-cell communication inference, and microscopy underscored the CXCL16-CXCR6 axis between KNIIFE and TRM cells. Secondary influenza challenge induced accelerated and coordinated myeloid and lymphoid responses without epithelial proliferation. Together, this atlas serves as a reference for viral infection in the upper respiratory tract and highlights the efficacy of local coordinated memory responses.
Collapse
Affiliation(s)
- Samuel W Kazer
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Immunology, Harvard Medical School, Boston, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Colette Matysiak Match
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Erica M Langan
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marie-Angèle Messou
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Thomas J LaSalle
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Program in Health Sciences and Technology, Harvard Medical School and Massachusetts Institute of Technology, Boston, MA, USA
| | - Elise O'Leary
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | | | | - Ulrich H von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Program in Immunology, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
5
|
Chen Z, Xiao L, Sun Q, Chen Q, Hua W, Zhang J. Effects of Acremonium terricola Culture on Lactation Performance, Immune Function, Antioxidant Capacity, and Intestinal Flora of Sows. Antioxidants (Basel) 2024; 13:970. [PMID: 39199216 PMCID: PMC11352107 DOI: 10.3390/antiox13080970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
This study aimed to determine the effects of different doses of Acremonium terricola culture (ATC) on lactation performance, immune function, antioxidant capacity, and intestinal flora of sows. Forty-five Landrace sows (3-6 parity) were randomly assigned to the following three treatments from 85 days of gestation to 21 days after farrowing: a control diet (CON, basal diet), a low-dose Acremonium terricola culture diet (0.2% ATC, basal diet + 0.2% ATC), and a high-dose Acremonium terricola culture diet (0.4% ATC, basal diet + 0.4% ATC). Compared with the CON group, the supplementation of 0.2% ATC increased the average daily milk yield of sows by 4.98%, increased milk fat, total solids, and freezing point depression on day 1 postpartum (p < 0.05), increased serum concentration of Triiodothyronine, Thyroxin, and Estradiol on day 21 postpartum (p < 0.05). Compared with the CON group, the supplementation of 0.4% ATC increased the average daily milk yield of sows by 9.38% (p < 0.05). Furthermore, the supplementation of 0.2% ATC increased serum concentration of IgG, IgM, and IFN-γ, CD4 on day 1 postpartum (p < 0.05) and increased serum concentration of immunoglobulin A ( IgA), immunoglobulin G (IgG), immunoglobulin M ( IgM), complement 3 (C3), cluster of differentiation 4 (CD4), cluster of differentiation 8 (CD8), interferon-γ (IFN-γ) on day 21 postpartum (p < 0.05), while the supplementation of 0.4% ATC reduced serum concentration of IL-2 on day 21 postpartum (p < 0.05). Moreover, the supplementation of 0.4% ATC significantly increased serum concentration of catalase (CAT) (p < 0.05). Additionally, the supplementation of ATC affected the relative abundance of the intestinal flora at different taxonomic levels in sows and increased the abundance of beneficial bacteria such as in the norank_f__Eubacterium_coprostanoligenes group, Eubacterium_coprostanoligenes group, and Lachnospiraceae_XPB1014 group of sows, while reducing the abundance of harmful bacteria such as Phascolarctobacterium and Clostridium_sensu_stricto_1. These data revealed that the supplementation of ATC during late gestation and lactation can improve lactation performance, immune function, antioxidant capacity, and the gut microbiota. Compared with supplementation of 0.4% ATC, 0.2% ATC enhances the levels of thyroid-related hormones, specific antibodies, and cytokines in serum, promotes the diversity of beneficial gut microbiota, beneficial bacteria in the intestine, reduces the population of harmful bacteria, and thereby bolsters the immunity of sows. Hence, 0.2% ATC is deemed a more optimal concentration.
Collapse
Affiliation(s)
- Zhirong Chen
- College of Animal Science, Zhejiang University, Hangzhou 310058, China; (Z.C.); (L.X.); (Q.S.); (Q.C.)
| | - Lixia Xiao
- College of Animal Science, Zhejiang University, Hangzhou 310058, China; (Z.C.); (L.X.); (Q.S.); (Q.C.)
| | - Qian Sun
- College of Animal Science, Zhejiang University, Hangzhou 310058, China; (Z.C.); (L.X.); (Q.S.); (Q.C.)
| | - Qiangqiang Chen
- College of Animal Science, Zhejiang University, Hangzhou 310058, China; (Z.C.); (L.X.); (Q.S.); (Q.C.)
| | - Weidong Hua
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China;
| | - Jinzhi Zhang
- College of Animal Science, Zhejiang University, Hangzhou 310058, China; (Z.C.); (L.X.); (Q.S.); (Q.C.)
| |
Collapse
|
6
|
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
|
7
|
Roberts LE, Williams CEC, Oni L, Barratt J, Selvaskandan H. IgA Nephropathy: Emerging Mechanisms of Disease. Indian J Nephrol 2024; 34:297-309. [PMID: 39156850 PMCID: PMC11326799 DOI: 10.25259/ijn_425_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 08/20/2024] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is the most common primary glomerulonephritis reported across the world and is characterized by immunoglobulin A (IgA) dominant mesangial deposits, which are poorly O-glycosylated. This deposition leads to a cascade of glomerular and tubulointerstitial inflammation and fibrosis, which can progress to chronic kidney disease. The variability in rate of progression reflects the many genetic and environmental factors that drive IgAN. Here, we summarize the contemporary understanding of the disease mechanisms that drive IgAN and provide an overview of new and emerging therapies, which target these mechanisms.
Collapse
Affiliation(s)
- Lydia E Roberts
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, United Kingdom
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Chloe E C Williams
- Royal Liverpool and Broadgreen University Hospital Trusts, Liverpool, United Kingdom
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Louise Oni
- Department of Women's and Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Paediatric Nephrology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, United Kingdom
| | - Jonathan Barratt
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, United Kingdom
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| | - Haresh Selvaskandan
- John Walls Renal Unit, University Hospitals of Leicester NHS Trust and University of Leicester, Leicester, United Kingdom
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
8
|
Kiekens C, Morré SA, Vanrompay D. Advances in Chlamydia trachomatis Vaccination: Unveiling the Potential of Major Outer Membrane Protein Derivative Constructs. Microorganisms 2024; 12:1196. [PMID: 38930578 PMCID: PMC11205628 DOI: 10.3390/microorganisms12061196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Chlamydia (C.) trachomatis, a leading cause of sexually transmitted infections (STIs) worldwide, continues to be a significant public health concern. The majority of infections are asymptomatic and, when left untreated, severe sequelae such as infertility and chronic pelvic pain can occur. Despite decades of research, an effective vaccine remains elusive. This review focuses on the potential of Major Outer Membrane Protein (MOMP)-derived constructs as promising candidates for C. trachomatis vaccination. MOMP, the most abundant protein in the outer membrane of C. trachomatis, has been a focal point of vaccine research over the years due to its antigenic properties. To overcome issues associated with the use of full MOMP as a vaccine antigen, derivative constructs have been studied. As these constructs are often not sufficiently immunogenic, antigen delivery systems or accompanying adjuvants are required. Additionally, several immunization routes have been explored with these MOMP-derived vaccine antigens, and determining the optimal route remains an ongoing area of research. Future directions and challenges in the field of C. trachomatis vaccination are discussed.
Collapse
Affiliation(s)
- Celien Kiekens
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Servaas A. Morré
- Department of Genetics and Cell Biology, GROW School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
- Microbe&Lab BV, 1105 AG Amsterdam, The Netherlands
- Dutch Chlamydia trachomatis Reference Laboratory, Department of Medical Microbiology, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Molecular and Cellular Engineering, Jacob Institute of Biotechnology and Bioengineering, Sam Higginbottom University of Agriculture, Technology and Sciences, Allahabad 211007, Uttar Pradesh, India
| | - Daisy Vanrompay
- Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
9
|
Etayo A, Bjørgen H, Hordvik I, Øvergård AC. Possible transport routes of IgM to the gut of teleost fish. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109583. [PMID: 38657879 DOI: 10.1016/j.fsi.2024.109583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/12/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024]
Abstract
Fish rely on mucosal surfaces as their first defence barrier against pathogens. Maintaining mucosal homeostasis is therefore crucial for their overall well-being, and it is likely that secreted immunoglobulins (sIg) play a pivotal role in sustaining this balance. In mammals, the poly-Ig receptor (pIgR) is an essential component responsible for transporting polymeric Igs across mucosal epithelia. In teleost fish, a counterpart of pIgR has been identified and characterized, exhibiting structural differences and broader mRNA expression patterns compared to mammals. Despite supporting evidence for the binding of Igs to recombinant pIgR proteins, the absence of a joining chain (J-chain) in teleosts challenges the conventional understanding of Ig transport mechanisms. The transport of IgM to the intestine via the hepatobiliary route is observed in vertebrates and has been proposed in a few teleosts. Investigations on the stomachless fish, ballan wrasse, revealed a significant role of the hepatobiliary route and interesting possibilities for alternative IgM transport routes that might include pancreatic tissue. These findings highlight the importance of gaining a thorough understanding of the mechanisms behind Ig transport to the gut in various teleosts. This review aims to gather existing information on pIgR-mediated transport across epithelial cells and immunoglobulin transport pathways to the gut lumen in teleost fish. It provides comparative insights into the hepatobiliary transport of Igs to the gut, emphasizing the current understanding in teleost fish while exploring potential alternative pathways for Ig transport to the gut lumen. Despite significant progress in understanding various aspects, there is still much to uncover, especially concerning the diversity of mechanisms across different teleost species.
Collapse
Affiliation(s)
- Angela Etayo
- Institute of Marine Research, Bergen, Norway; Fish Health group, Department of Biological sciences, University of Bergen, Norway.
| | - Håvard Bjørgen
- Anatomy Unit, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ås, Norway
| | - Ivar Hordvik
- Fish Health group, Department of Biological sciences, University of Bergen, Norway
| | | |
Collapse
|
10
|
Kazer SW, Match CM, Langan EM, Messou MA, LaSalle TJ, O’Leary E, Marbourg J, Naughton K, von Andrian UH, Ordovas-Montanes J. Primary nasal viral infection rewires the tissue-scale memory response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.05.11.539887. [PMID: 38562902 PMCID: PMC10983857 DOI: 10.1101/2023.05.11.539887] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The nasal mucosa is frequently the initial site of respiratory viral infection, replication, and transmission. Recent work has started to clarify the independent responses of epithelial, myeloid, and lymphoid cells to viral infection in the nasal mucosa, but their spatiotemporal coordination and relative contributions remain unclear. Furthermore, understanding whether and how primary infection shapes tissue-scale memory responses to secondary challenge is critical for the rational design of nasal-targeting therapeutics and vaccines. Here, we generated a single-cell RNA-sequencing (scRNA-seq) atlas of the murine nasal mucosa sampling three distinct regions before and during primary and secondary influenza infection. Primary infection was largely restricted to respiratory mucosa and induced stepwise changes in cell type, subset, and state composition over time. Type I Interferon (IFN)-responsive neutrophils appeared 2 days post infection (dpi) and preceded transient IFN-responsive/cycling epithelial cell responses 5 dpi, which coincided with broader antiviral monocyte and NK cell accumulation. By 8 dpi, monocyte-derived macrophages (MDMs) expressing Cxcl9 and Cxcl16 arose alongside effector cytotoxic CD8 and Ifng-expressing CD4 T cells. Following viral clearance (14 dpi), rare, previously undescribed Krt13+ nasal immune-interacting floor epithelial (KNIIFE) cells expressing multiple genes with immune communication potential increased concurrently with tissue-resident memory T (TRM)-like cells and early IgG+/IgA+ plasmablasts. Proportionality analysis coupled with cell-cell communication inference, alongside validation by in situ microscopy, underscored the CXCL16-CXCR6 signaling axis between MDMs and effector CD8 T cells 8dpi and KNIIFE cells and TRM cells 14 dpi. Secondary influenza challenge with a homologous or heterologous strain administered 60 dpi induced an accelerated and coordinated myeloid and lymphoid response without epithelial proliferation, illustrating how tissue-scale memory to natural infection engages both myeloid and lymphoid cells to reduce epithelial regenerative burden. Together, this atlas serves as a reference for viral infection in the upper respiratory tract and highlights the efficacy of local coordinated memory responses upon rechallenge.
Collapse
Affiliation(s)
- Samuel W. Kazer
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Colette Matysiak Match
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Erica M. Langan
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marie-Angèle Messou
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Thomas J. LaSalle
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA
| | - Elise O’Leary
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | | | | - Ulrich H. von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
11
|
Castro VT, Chardin H, Amorim dos Santos J, Barra GB, Castilho GR, Souza PM, Magalhães PDO, Acevedo AC, Guerra ENS. Detection of anti-SARS-CoV-2 salivary antibodies in vaccinated adults. Front Immunol 2023; 14:1296603. [PMID: 38022522 PMCID: PMC10661372 DOI: 10.3389/fimmu.2023.1296603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Since the introduction of efficient anti-SARS-CoV-2 vaccines, the detection of antibodies becomes useful for immunological monitoring and COVID-19 control. Therefore, this longitudinal study aimed to evaluate the detection of SARS-CoV-2 antibodies in the serum and saliva of COVID-19-vaccinated adults. The study included 13 not vaccinated and 35 vaccinated participants with two doses of CoronaVac (Sinovac/Butantan) vaccine who subsequently received BNT162b2 (Pfizer-BioNTech) vaccine as a booster dose. Vaccinated participants donated saliva and serum in three different time points. Enzyme-linked immunosorbent assay was used for antibody detection. In our results, the serum neutralizing antibodies (NAb) were detected in 34/35 samples after second dose and in 35/35 samples one and five months after the booster dose. In saliva, NAb were detected in 30/35 samples after second dose and in 35/35 of samples one and five months after the booster dose. IgA was detected in 19/34 saliva samples after second dose, in 18/35 one month after the booster and in 30/35 five months after. IgG in saliva was detected in 1/34 samples after second dose, 33/35 samples one month after the booster dose and in 20/35 five months after. A strong correlation was found between IgG and neutralizing activity in saliva, and salivary IgA would be a sign of recent exposure to the virus. In conclusion, saliva can be suitable for monitoring antibodies anti-SARS-CoV-2 after vaccination. Heterologous vaccination contributed to increase anti-SARS-CoV-2 antibodies in the Brazilian health context. Complementary studies with large groups are mandatory to conclude the interest in following mucosal immunity.
Collapse
Affiliation(s)
- Vitória Tavares Castro
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Hélène Chardin
- Department of Analytical, Bioanalytical Sciences and Miniaturization, École Supérieure de Physique et de Chimie Industrielles (ESPCI) de la Ville de Paris, Paris, France
- Unité de Formation et de Recherche d’Odontologie, Université Paris Cité, Paris, France
| | - Juliana Amorim dos Santos
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | | | | | - Paula Monteiro Souza
- Laboratory of Natural Products, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | | | - Ana Carolina Acevedo
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| | - Eliete Neves Silva Guerra
- Laboratory of Oral Histopathology, Faculty of Health Sciences, University of Brasilia, Brasília, DF, Brazil
| |
Collapse
|
12
|
Kabue JP, Khumela R, Meader E, Baroni de Moraes MT, Traore AN, Potgieter N. Norovirus-Associated Gastroenteritis Vesikari Score and Pre-Existing Salivary IgA in Young Children from Rural South Africa. Viruses 2023; 15:2185. [PMID: 38005863 PMCID: PMC10674611 DOI: 10.3390/v15112185] [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] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Norovirus (NoV) is the leading cause of viral gastroenteritis, mostly affecting young children worldwide. However, limited data are available to determine the severity of norovirus-associated AGE (acute gastroenteritis) and to correlate it with the NoV-specific IgA antibodies' level. Between October 2019 and September 2021, two hundred stool samples were randomly collected from symptomatic cases for the vesikari score and NoV-specific IgA assessment in young children from rural South Africa. Additionally, one hundred saliva specimens were concomitantly sampled within the same cohort to evaluate the NoV-specific salivary IgA levels. In addition, 50 paired saliva and stool samples were simultaneously collected from asymptomatic children to serve as controls. NoV strains in stool samples were detected using real-time RT-PCR, amplified, and genotyped with RT-PCR and Sanger sequencing. ELISA using NoV VLP (virus-like particles) GII.4 as antigens was performed on the saliva specimens. Dehydrated children were predominantly those with NoV infections (65/74, 88%; p < 0.0001). NoV-positive infections were significantly associated with the severe diarrhea cases having a high vesikari score (55%, 33/60) when compared to the non-severe diarrheal score (29.3%, 41/140; p < 0.0308). NoV of the GII genogroup was mainly detected in severe diarrhea cases (50.9%, 30/59; p = 0.0036). The geometric means of the NoV-specific IgA level were higher in the asymptomatic NoV-infected group (0.286) as compared to the symptomatic group (0.174). This finding suggests that mucosal immunity may not protect the children from the NoV infection. However, the findings indicated the contribution of the pre-existing NoV-specific IgA immune response in reducing the severity of diarrheal disease. A high vesikari score of AGE associated with the NoV GII genogroup circulating in the study area underscores the need for an appropriate treatment of AGE based on the severity level of NoV-associated clinical symptoms in young children.
Collapse
Affiliation(s)
- Jean-Pierre Kabue
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Private Bag X5050, Thohoyandou 0950, South Africa; (R.K.); (A.N.T.); (N.P.)
| | - Ronewa Khumela
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Private Bag X5050, Thohoyandou 0950, South Africa; (R.K.); (A.N.T.); (N.P.)
| | - Emma Meader
- Clinical Microbiology, Pathology Department, East Kent Hospitals University NHS Foundation Trust, Ashford TN24 OLZ, UK;
| | - Marcia Terezinha Baroni de Moraes
- Laboratory of Comparative and Environmental Virology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Avenida Brazil, 4365-Manguinhos, Rio de Janeiro 21040-360, RJ, Brazil;
| | - Afsatou Ndama Traore
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Private Bag X5050, Thohoyandou 0950, South Africa; (R.K.); (A.N.T.); (N.P.)
| | - Natasha Potgieter
- Department of Biochemistry and Microbiology, Faculty of Science, Engineering and Agriculture, University of Venda, Private Bag X5050, Thohoyandou 0950, South Africa; (R.K.); (A.N.T.); (N.P.)
| |
Collapse
|
13
|
Lorgen-Ritchie M, Uren Webster T, McMurtrie J, Bass D, Tyler CR, Rowley A, Martin SAM. Microbiomes in the context of developing sustainable intensified aquaculture. Front Microbiol 2023; 14:1200997. [PMID: 37426003 PMCID: PMC10327644 DOI: 10.3389/fmicb.2023.1200997] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
With an ever-growing human population, the need for sustainable production of nutritional food sources has never been greater. Aquaculture is a key industry engaged in active development to increase production in line with this need while remaining sustainable in terms of environmental impact and promoting good welfare and health in farmed species. Microbiomes fundamentally underpin animal health, being a key part of their digestive, metabolic and defense systems, in the latter case protecting against opportunistic pathogens in the environment. The potential to manipulate the microbiome to the advantage of enhancing health, welfare and production is an intriguing prospect that has gained considerable traction in recent years. In this review we first set out what is known about the role of the microbiome in aquaculture production systems across the phylogenetic spectrum of cultured animals, from invertebrates to finfish. With a view to reducing environmental footprint and tightening biological and physical control, investment in "closed" aquaculture systems is on the rise, but little is known about how the microbial systems of these closed systems affect the health of cultured organisms. Through comparisons of the microbiomes and their dynamics across phylogenetically distinct animals and different aquaculture systems, we focus on microbial communities in terms of their functionality in order to identify what features within these microbiomes need to be harnessed for optimizing healthy intensified production in support of a sustainable future for aquaculture.
Collapse
Affiliation(s)
| | - Tamsyn Uren Webster
- Centre for Sustainable Aquatic Research, Swansea University, Swansea, United Kingdom
| | - Jamie McMurtrie
- College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - David Bass
- College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
- Centre for Environment, Fisheries and Aquaculture Science (Cefas), Weymouth, United Kingdom
| | - Charles R. Tyler
- College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Andrew Rowley
- Department of Biosciences, Faculty of Science and Engineering, Swansea University, Swansea, United Kingdom
| | - Samuel A. M. Martin
- School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
14
|
Grygiel-Górniak B, Folga BA. Chlamydia trachomatis-An Emerging Old Entity? Microorganisms 2023; 11:1283. [PMID: 37317257 DOI: 10.3390/microorganisms11051283] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/04/2023] [Accepted: 05/12/2023] [Indexed: 06/16/2023] Open
Abstract
Chlamydia trachomatis is an evasive pathogen that can prompt severe clinical manifestations in humans such as vaginitis, epididymitis, lymphogranuloma venereum, trachoma, conjunctivitis and pneumonia. If left untreated, chronic infections with C. trachomatis can give rise to long-lasting and even permanent sequelae. To shed some light on its widespread nature, data from original research, systematic reviews and meta-analyses from three databases was collected and analyzed in the context of chlamydial infection, related symptoms and appropriate treatment modalities. This review describes the bacterium's pervasiveness on a global scale, especially in developing countries, and suggests ways to halt its transmission and spread. Infections with C. trachomatis often go unnoticed, as many individuals are asymptomatic and unaware of their diagnosis, contributing to a delay in diagnosis and treatment. The high prevalence of chlamydial infection highlights the need for a universal screening and detection method enabling immediate treatment at its onset. Prognosis is favorable with antibiotic therapy and education for high-risk groups and their sexual partners. In the future, a quick, easily accessible, and inexpensive test should be developed to diagnose and treat infected individuals early on. Along with a vaccine against C. trachomatis, it would halt the transmission and spread of the pathogen worldwide.
Collapse
Affiliation(s)
- Bogna Grygiel-Górniak
- Department of Rheumatology, Rehabilitation and Internal Diseases, Poznan University of Medical Sciences, 61-701 Poznań, Poland
| | - Barbara Anna Folga
- Department of Rheumatology, Rehabilitation and Internal Diseases, Poznan University of Medical Sciences, 61-701 Poznań, Poland
| |
Collapse
|
15
|
Kok TW, Izzo AA, Costabile M. Intracellular immunoglobulin A (icIgA) in protective immunity and vaccines. Scand J Immunol 2023; 97:e13253. [PMID: 36597220 DOI: 10.1111/sji.13253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/20/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023]
Abstract
Virus neutralization at respiratory mucosal surfaces is important in the prevention of infection. Mucosal immunity is mediated mainly by extracellular secretory immunoglobulin A (sIgA) and its role has been well studied. However, the protective role of intracellular specific IgA (icIgA) is less well defined. Initially, in vitro studies using epithelial cell lines with surface expressed polymeric immunoglobulin receptor (pIgR) in transwell culture chambers have shown that icIgA can neutralize influenza, parainfluenza, HIV, rotavirus and measles viruses. This effect appears to involve an interaction between polymeric immunoglobulin A (pIgA) and viral particles within an intracellular compartment, since IgA is transported across the polarized cell. Co-localization of specific icIgA with influenza virus in patients' (virus culture positive) respiratory epithelial cells using well-characterized antisera was initially reported in 2018. This review provides a summary of in vitro studies with icIgA on colocalization and neutralization of the above five viruses. Two other highly significant respiratory infectious agents with severe global impacts viz. SARS-2 virus (CoViD pandemic) and the intracellular bacterium-Mycobacterium tuberculosis-are discussed. Further studies will provide more detailed understanding of the mechanisms and kinetics of icIgA neutralization in relation to viral entry and early replication steps with a specific focus on mucosal infections. This will inform the design of more effective vaccines against infectious agents transmitted via the mucosal route.
Collapse
Affiliation(s)
- Tuck-Weng Kok
- University of Adelaide, Faculty of Health & Medical Sciences and School of Biological Sciences, Adelaide, South Australia, Australia
| | - Angelo A Izzo
- University of Sydney, Tuberculosis Research Program, Centenary Institute, Camperdown, New South Wales, Australia
| | - Maurizio Costabile
- University of South Australia, Clinical and Health Sciences and Centre for Cancer Biology, Adelaide, South Australia, Australia
| |
Collapse
|
16
|
Sun L, Zhao N, Li H, Wang B, Li H, Zhang X, Zhao X. Construction of a Lactobacillus plantarum-based claudin-3 targeting delivery system for the development of vaccines against Eimeria tenella. Vaccine 2023; 41:756-765. [PMID: 36526500 DOI: 10.1016/j.vaccine.2022.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/06/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Avian coccidiosis causes huge economic losses to the poultry industry worldwide and currently lacks effective live vector vaccines. Achieving efficient antigen delivery to mucosa-associated lymphoid tissue (MALT) is critical for improving the effectiveness of vaccines. Here, chicken claudin-3 (CLDN3), a tight junction protein expressed in MALT, was identified as a target, and the C-terminal region of Clostridium perfringens enterotoxin (C-CPE) was proven to bind to chicken CLDN3. Then, a CLDN3-targeting Lactobacillus plantarum NC8-expressing C-CPE surface display system (NC8/GFP-C-CPE) was constructed to successfully express the heterologous protein on the surface of L. plantarum. The colonization level of NC8/GFP-C-CPE was significantly increased compared to the non-targeting strain and could persist in the intestine for at least 72 h. An oral vaccine strain expressing five EGF domains of Eimeria tenella microneme protein 8 (EtMIC8-EGF) (NC8/EtMIC8-EGF-C-CPE) was constructed to evaluate the protective efficacy against E. tenella infection. The results revealed that CLDN3-targeting L. plantarum induced stronger mucosal immunity in gut-associated lymphoid tissues (GALT) as well as humoral responses and conferred better protection in terms of parasite replication and pathology than the non-targeting strain. Overall, we successfully constructed a CLDN3-targeting L. plantarum NC8 surface display system characterized by MALT-targeting, which is an efficient antigen delivery system to confer enhanced protective efficacy in chickens against E. tenella infection.
Collapse
Affiliation(s)
- Lingyu Sun
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Ningning Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Huihui Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Bingxiang Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Hongmei Li
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China
| | - Xiao Zhang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China.
| | - Xiaomin Zhao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Tai'an City, Shandong Province, China; Shandong Provincial Engineering Technology Research Center of Animal Disease Control and Prevention, Shandong Agricultural University, 61 Daizong Street, Tai'an City 271018, Shandong Province, China.
| |
Collapse
|
17
|
Tsai CJY, Loh JMS, Fujihashi K, Kiyono H. Mucosal vaccination: onward and upward. Expert Rev Vaccines 2023; 22:885-899. [PMID: 37817433 DOI: 10.1080/14760584.2023.2268724] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION The unique mucosal immune system allows the generation of robust protective immune responses at the front line of pathogen encounters. The needle-free delivery route and cold chain-free logistic requirements also provide additional advantages in ease and economy. However, the development of mucosal vaccines faces several challenges, and only a handful of mucosal vaccines are currently licensed. These vaccines are all in the form of live attenuated or inactivated whole organisms, whereas no subunit-based mucosal vaccine is available. AREAS COVERED The selection of antigen, delivery vehicle, route and adjuvants for mucosal vaccination are highly important. This is particularly crucial for subunit vaccines, as they often fail to elicit strong immune responses. Emerging research is providing new insights into the biological and immunological uniqueness of mucosal tissues. However, many aspects of the mucosal immunology still await to be investigated. EXPERT OPINION This article provides an overview of the current understanding of mucosal vaccination and discusses the remaining knowledge gaps. We emphasize that because of the potential benefits mucosal vaccines can bring from the biomedical, social and economic standpoints, the unmet goal to achieve mucosal vaccine success is worth the effort.
Collapse
Affiliation(s)
- Catherine J Y Tsai
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
| | - Jacelyn M S Loh
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hiroshi Kiyono
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
- CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
- Future Medicine Education and Research Organization, Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan
| |
Collapse
|
18
|
Romero-Ramírez S, Sosa-Hernández VA, Cervantes-Díaz R, Carrillo-Vázquez DA, Meza-Sánchez DE, Núñez-Álvarez C, Torres-Ruiz J, Gómez-Martín D, Maravillas-Montero JL. Salivary IgA subtypes as novel disease biomarkers in systemic lupus erythematosus. Front Immunol 2023; 14:1080154. [PMID: 36911711 PMCID: PMC9992540 DOI: 10.3389/fimmu.2023.1080154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/13/2023] [Indexed: 02/24/2023] Open
Abstract
Introduction Immunoglobulin A (IgA) is the main antibody isotype in body fluids such as tears, intestinal mucous, colostrum, and saliva. There are two subtypes of IgA in humans: IgA1, mainly present in blood and mucosal sites, and IgA2, preferentially expressed in mucosal sites like the colon. In clinical practice, immunoglobulins are typically measured in venous or capillary blood; however, alternative samples, including saliva, are now being considered, given their non-invasive and easy collection nature. Several autoimmune diseases have been related to diverse abnormalities in oral mucosal immunity, such as rheumatoid arthritis, Sjogren's syndrome, and systemic lupus erythematosus (SLE). Methods We decided to evaluate the levels of both IgA subtypes in the saliva of SLE patients. A light chain capture-based ELISA measured specific IgA1 and IgA2 levels in a cohort of SLE patients compared with age and gender-matched healthy volunteers. Results Surprisingly, our results indicated that in the saliva of SLE patients, total IgA and IgA1 subtype were significantly elevated; we also found that salivary IgA levels, particularly IgA2, positively correlate with anti-dsDNA IgG antibody titers. Strikingly, we also detected the presence of salivary anti-nucleosome IgA antibodies in SLE patients, a feature not previously reported elsewhere. Conclusions According to our results and upon necessary validation, IgA characterization in saliva could represent a potentially helpful tool in the clinical care of SLE patients with the advantage of being a more straightforward, faster, and safer method than manipulating blood samples.
Collapse
Affiliation(s)
- Sandra Romero-Ramírez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Víctor A Sosa-Hernández
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Rodrigo Cervantes-Díaz
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Daniel A Carrillo-Vázquez
- Departamento de Medicina Interna, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - David E Meza-Sánchez
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Carlos Núñez-Álvarez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jiram Torres-Ruiz
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Gómez-Martín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José L Maravillas-Montero
- Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México e Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
19
|
Ghasemi A, Wang S, Sahay B, Abbott JR, Curtiss R. Protective immunity enhanced Salmonella vaccine vectors delivering Helicobacter pylori antigens reduce H. pylori stomach colonization in mice. Front Immunol 2022; 13:1034683. [PMID: 36466847 PMCID: PMC9716130 DOI: 10.3389/fimmu.2022.1034683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2024] Open
Abstract
Helicobacter pylori is a major cause of gastric mucosal inflammation, peptic ulcers, and gastric cancer. Emerging antimicrobial-resistant H. pylori has hampered the effective eradication of frequent chronic infections. Moreover, a safe vaccine is highly demanded due to the absence of effective vaccines against H. pylori. In this study, we employed a new innovative Protective Immunity Enhanced Salmonella Vaccine (PIESV) vector strain to deliver and express multiple H. pylori antigen genes. Immunization of mice with our vaccine delivering the HpaA, Hp-NAP, UreA and UreB antigens, provided sterile protection against H. pylori SS1 infection in 7 out of 10 tested mice. In comparison to the control groups that had received PBS or a PIESV carrying an empty vector, immunized mice exhibited specific and significant cellular recall responses and antigen-specific serum IgG1, IgG2c, total IgG and gastric IgA antibody titers. In conclusion, an improved S. Typhimurium-based live vaccine delivering four antigens shows promise as a safe and effective vaccine against H. pylori infection.
Collapse
Affiliation(s)
- Amir Ghasemi
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Shifeng Wang
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Bikash Sahay
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| | - Jeffrey R. Abbott
- Department of Comparative, Diagnostic and Population Medicine, University of Florida, Gainesville, FL, United States
| | - Roy Curtiss
- Department of Infectious Diseases and Immunology, College of Veterinary Medicine, University of Florida, Gainesville, Florida, FL, United States
| |
Collapse
|
20
|
Beneficial health effects of polyphenols metabolized by fermentation. Food Sci Biotechnol 2022; 31:1027-1040. [DOI: 10.1007/s10068-022-01112-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/09/2022] [Accepted: 05/29/2022] [Indexed: 11/04/2022] Open
|
21
|
Alenquer M, Milheiro Silva T, Akpogheneta O, Ferreira F, Vale-Costa S, Medina-Lopes M, Batista F, Garcia AM, Barreto VM, Paulino C, Costa J, Sobral J, Diniz-da-Costa M, Ladeiro S, Corte-Real R, Delgado Alves J, Leite RB, Demengeot J, Rocha Brito MJ, Amorim MJ. Saliva molecular testing bypassing RNA extraction is suitable for monitoring and diagnosing SARS-CoV-2 infection in children. PLoS One 2022; 17:e0268388. [PMID: 35704567 PMCID: PMC9200166 DOI: 10.1371/journal.pone.0268388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/28/2022] [Indexed: 01/08/2023] Open
Abstract
Background Adults are being vaccinated against SARS-CoV-2 worldwide, but the longitudinal protection of these vaccines is uncertain, given the ongoing appearance of SARS-CoV-2 variants. Children remain largely unvaccinated and are susceptible to infection, with studies reporting that they actively transmit the virus even when asymptomatic, thus affecting the community. Methods We investigated if saliva is an effective sample for detecting SARS-CoV-2 RNA and antibodies in children, and associated viral RNA levels to infectivity. For that, we used a saliva-based SARS-CoV-2 RT-qPCR test, preceded or not by RNA extraction, in 85 children aged 10 years and under, admitted to the hospital regardless of COVID-19 symptomatology. Amongst these, 29 (63.0%) presented at least one COVID-19 symptom, 46 (54.1%) were positive for SARS-CoV-2 infection, 28 (32.9%) were under the age of 1, and the mean (SD) age was 3.8 (3.4) years. Saliva samples were collected up to 48 h after a nasopharyngeal swab-RT-qPCR test. Results In children aged 10 years and under, the sensitivity, specificity, and accuracy of saliva-RT-qPCR tests compared to NP swab-RT-qPCR were, respectively, 84.8% (71.8%–92.4%), 100% (91.0%–100%), and 91.8% (84.0%–96.6%) with RNA extraction, and 81.8% (68.0%–90.5%), 100% (91.0%–100%), and 90.4% (82.1%–95.0%) without RNA extraction. Rescue of infectious particles from saliva was limited to CT values below 26. In addition, we found significant IgM positive responses to SARS-CoV-2 in children positive for SARS-CoV-2 by NP swab and negative by saliva compared to other groups, indicating late infection onset (>7–10 days). Conclusions Saliva is a suitable sample type for diagnosing children aged 10 years and under, including infants aged <1 year, even bypassing RNA extraction methods. Importantly, the detected viral RNA levels were significantly above the infectivity threshold in several samples. Further investigation is required to correlate SARS-CoV-2 RNA levels to viral transmission.
Collapse
Affiliation(s)
- Marta Alenquer
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Tiago Milheiro Silva
- Pediatric Infectious Disease Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal
| | - Onome Akpogheneta
- Lymphocyte Physiology Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Filipe Ferreira
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Sílvia Vale-Costa
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Mónica Medina-Lopes
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Frederico Batista
- Department of Medicine 4, Hospital Prof Doutor Fernando Fonseca, Amadora, Portugal
| | - Ana Margarida Garcia
- Pediatric Infectious Disease Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal
| | - Vasco M. Barreto
- CEDOC NOVA, Centro de Estudos de Doenças Crónicas, Nova Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Cathy Paulino
- Genomics Unit, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - João Costa
- Genomics Unit, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - João Sobral
- Genomics Unit, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | | - Susana Ladeiro
- Genomics Unit, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Rita Corte-Real
- Molecular Biology Laboratory, Department of Clinical Pathology, Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal
| | - José Delgado Alves
- Department of Medicine 4, Hospital Prof Doutor Fernando Fonseca, Amadora, Portugal
- CEDOC NOVA, Centro de Estudos de Doenças Crónicas, Nova Medical School, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Ricardo B. Leite
- Genomics Unit, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Jocelyne Demengeot
- Lymphocyte Physiology Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Maria João Rocha Brito
- Pediatric Infectious Disease Unit, Hospital Dona Estefânia, Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
- * E-mail:
| |
Collapse
|
22
|
Sheng X, Guo Y, Zhu H, Chai B, Tang X, Xing J, Chi H, Zhan W. Transepithelial Secretion of Mucosal IgM Mediated by Polymeric Immunoglobulin Receptor of Flounder ( Paralichthys olivaceus): In-Vivo and In-Vitro Evidence. Front Immunol 2022; 13:868753. [PMID: 35464454 PMCID: PMC9019723 DOI: 10.3389/fimmu.2022.868753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/14/2022] [Indexed: 12/02/2022] Open
Abstract
Secretory immunoglobulin (SIg) is crucial for mucosal surface defenses, but the transepithelial secretion of SIg mediated by polymeric immunoglobulin receptor (pIgR) is not clarified in fish. We previously found that flounder (Paralichthys olivaceus) pIgR (fpIgR) and secretory IgM (SIgM) increased in gut mucus post-vaccination. Here, the fpIgR-positive signal was mainly observed in the intestinal epithelium, whereas the IgM-positive signal was mainly distributed in the lamina propria, before immunization. IgM signals increased in the lamina propria and then in the epithelium after immunization with inactivated Vibrio anguillarum, and co-localization between IgM and fpIgR in the epithelium was determined, while the presence of EdU+IgM+ cells in the lamina propria identified the proliferative B cells, revealing that the secretion and transepithelial transport of SIgM locally occurred in the gut of flounder. Subsequently, we established an in-vitro model of transfected MDCK cells that stably expressed the fpIgR. After a recombinant eukaryotic expression plasmid (pCIneoEGFP-fpIgR) was constructed and transfected into MDCK cells, stable expression of the fpIgR in transfected MDCK-fpIgR cells was confirmed, and the tightness and integrity of the polarized cell monolayers grown on Transwells were evaluated. Afterward, the serum IgM of flounder was purified as a binding ligand and placed in the lower compartment of Transwells. An ~800-kDa protein band in the upper compartment was shown to be IgM- and fpIgR-positive, and IgM-positive fluorescence was seen in MDCK-fpIgR cells but not in MDCK-mock cells. Hence, the fpIgR helped polymeric IgM to pass across MDCK-fpIgR cells via transcytosis in a basolateral-to-apical fashion. These new findings provide a better understanding of the pathways shaping mucosal IgM responses and the local mucosal immune mechanisms in teleosts.
Collapse
Affiliation(s)
- Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture of Ministry of Education (KLMME), Ocean University of China, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yuan Guo
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture of Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Hui Zhu
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture of Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Baihui Chai
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture of Ministry of Education (KLMME), Ocean University of China, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture of Ministry of Education (KLMME), Ocean University of China, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture of Ministry of Education (KLMME), Ocean University of China, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture of Ministry of Education (KLMME), Ocean University of China, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, Key Laboratory of Mariculture of Ministry of Education (KLMME), Ocean University of China, Qingdao, China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
23
|
Boussamet L, Rajoka MSR, Berthelot L. Microbiota, IgA and Multiple Sclerosis. Microorganisms 2022; 10:microorganisms10030617. [PMID: 35336190 PMCID: PMC8954136 DOI: 10.3390/microorganisms10030617] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/11/2022] [Accepted: 03/11/2022] [Indexed: 12/16/2022] Open
Abstract
Multiple sclerosis (MS) is a neuroinflammatory disease characterized by immune cell infiltration in the central nervous system and destruction of myelin sheaths. Alterations of gut bacteria abundances are present in MS patients. In mouse models of neuroinflammation, depletion of microbiota results in amelioration of symptoms, and gavage with MS patient microbiota exacerbates the disease and inflammation via Th17 cells. On the other hand, depletion of B cells using anti-CD20 is an efficient therapy in MS, and growing evidence shows an important deleterious role of B cells in MS pathology. However, the failure of TACI-Ig treatment in MS highlighted the potential regulatory role of plasma cells. The mechanism was recently demonstrated involving IgA+ plasma cells, specific for gut microbiota and producing IL-10. IgA-coated bacteria in MS patient gut exhibit also modifications. We will focus our review on IgA interactions with gut microbiota and IgA+ B cells in MS. These recent data emphasize new pathways of neuroinflammation regulation in MS.
Collapse
Affiliation(s)
- Léo Boussamet
- Centre for Research in Transplantation and Translation Immunology, Nantes Université, Inserm, CR2TI UMR, 1064 Nantes, France;
| | - Muhammad Shahid Riaz Rajoka
- Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan;
| | - Laureline Berthelot
- Centre for Research in Transplantation and Translation Immunology, Nantes Université, Inserm, CR2TI UMR, 1064 Nantes, France;
- Correspondence:
| |
Collapse
|
24
|
Orally Administrated Recombinant Vaccinia Virus Displaying ROP4 Induces Protection against Toxoplasma gondii Challenge Infection. Vaccines (Basel) 2022; 10:vaccines10020152. [PMID: 35214611 PMCID: PMC8878533 DOI: 10.3390/vaccines10020152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 11/16/2022] Open
Abstract
Recombinant vaccinia viruses (rVVs) are attenuated viruses and are widely utilized as vectored vaccine platforms against numerous diseases. However, the protective efficacy of these rVV vaccines against Toxoplasma gondii and the resulting mucosal immunity has not been thoroughly assessed. Here, rVVs expressing the rhoptry protein 4 (ROP4) of T. gondii were generated. To evaluate the protection induced by the vaccines, mice were orally immunized with the ROP4-rVVs and subsequently challenge-infected with a lethal dose of T. gondii ME49 strain. Immunization with the rVVs induced higher levels of parasite-specific IgG and IgA antibody responses in sera compared to unimmunized control (NC). Upon challenge infection, significantly higher levels of IgG or IgA antibody responses in the brain, intestines, and vaginal samples were found in the immunized mice compared to NC. The ROP4-rVV vaccination elicited potent IgG and IgA secreting cell (ASC) responses, while substantially enhancing germinal center B cell, as well as CD4+ and CD8+ T cell responses from lymphoid organs. The production of pro-inflammatory cytokines IFN-γ and IL-6 in the brains was markedly diminished following immunization. The immunized mice also experienced reduced bodyweight loss and possessed fewer brain cysts than the control group. These results suggest that oral delivery of ROP4 displaying rVVs induced mucosal and systemic immunities that contributed to protection against lethal T. gondii challenge infection.
Collapse
|
25
|
Zorgi NE, Meireles LR, Oliveira DBL, Araujo DB, Durigon EL, Andrade Junior HFD. Isolated specific IgA against respiratory viruses, Influenza or SARS-CoV-2, present in the saliva of a fraction of healthy and asymptomatic volunteers. Clinics (Sao Paulo) 2022; 77:100105. [PMID: 36116267 PMCID: PMC9444893 DOI: 10.1016/j.clinsp.2022.100105] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 06/07/2022] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Defense against respiratory viruses depends on an immune response present in the mucosa, as saliva IgA secretes antibodies. During the pandemic, such as influenza or SARS-CoV-2, most infected patients are asymptomatic but retain specific antibodies post-infection. The authors evaluated IgG and IgA antibodies against SARS-CoV-2 and influenza in the saliva of asymptomatic volunteers, validated with controls or vaccinated individuals. METHODS The authors detected specific antibodies by validated conventional ELISA using natural SARS-CoV-2 antigens from infected Vero cells or capture-ELISA for influenza using natural antigens of the influenza vaccine. RESULTS Saliva from influenza-vaccinated individuals had more IgA than paired serum, contrary to the findings for specific IgG. In COVID-19-vaccinated samples, specific IgA in saliva increased after vaccination, but IgG levels were high after the first dose. In saliva from the asymptomatic population (226), anti-Influenza IgG was found in 57.5% (130) of samples, higher than IgA, found in 35% (79) of samples. IgA results were similar for SARS-CoV-2, with IgA present in 30% (68) of samples, while IgG was less present, in 44.2% (100) of samples. The proportion of influenza IgG responders was higher than that for SARS-CoV-2 IgG, but both populations presented similar proportions of IgA responders, possibly due to variable memory B cell survival. For both viruses, the authors found an important proportion (> 10%) of IgA+IgG- samples, suggesting the occurrence of humoral immunity directed to the mucosa. CONCLUSION Specific antibodies for respiratory viruses in saliva are found in either infection or vaccination and are a convenient and sensitive diagnostic tool for host immune response.
Collapse
Affiliation(s)
- Nahiara Esteves Zorgi
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil
| | - Luciana R Meireles
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil
| | | | - Danielle Bastos Araujo
- Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Edson L Durigon
- Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Heitor Franco de Andrade Junior
- Faculdade de Medicina da Universidade São Paulo (FMUSP), São Paulo, SP, Brazil; Laboratório de Protozoologia, Instituto de Medicina Tropical de São Paulo (IMTSP), São Paulo, SP, Brazil.
| |
Collapse
|
26
|
Gonçalves J, Juliano AM, Charepe N, Alenquer M, Athayde D, Ferreira F, Archer M, Amorim MJ, Serrano F, Soares H. Secretory IgA and T cells targeting SARS-CoV-2 spike protein are transferred to the breastmilk upon mRNA vaccination. Cell Rep Med 2021; 2:100468. [PMID: 34873588 PMCID: PMC8636305 DOI: 10.1016/j.xcrm.2021.100468] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/09/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022]
Abstract
In view of the scarcity of data to guide decision making, we evaluated how BNT162b2 and mRNA-1273 vaccines affect the immune response in lactating women and the protective profile of breastmilk. Compared with controls, lactating women had a higher frequency of circulating RBD memory B cells and higher anti-RBD antibody titers but similar neutralizing capacity. We show that upon vaccination, immune transfer to breastmilk occurs through a combination of anti-spike secretory IgA (SIgA) antibodies and spike-reactive T cells. Although we found that the concentration of anti-spike IgA in breastmilk might not be sufficient to directly neutralize SARS-CoV-2, our data suggest that cumulative transfer of IgA might provide the infant with effective neutralization capacity. Our findings put forward the possibility that breastmilk might convey both immediate (through anti-spike SIgA) and long-lived (via spike-reactive T cells) immune protection to the infant. Further studies are needed to address this possibility and to determine the functional profile of spike T cells.
Collapse
Affiliation(s)
- Juliana Gonçalves
- Human Immunobiology and Pathogenesis Group, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
- iNOVA4Health, Lisbon, Portugal
| | - A. Margarida Juliano
- Human Immunobiology and Pathogenesis Group, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
- iNOVA4Health, Lisbon, Portugal
| | - Nádia Charepe
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
| | - Marta Alenquer
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Diogo Athayde
- Membrane Protein Crystallography Laboratory, Instituto de Tecnologia Química e Biológica, ITQB-NOVA, Oeiras, Portugal
| | - Filipe Ferreira
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Margarida Archer
- Membrane Protein Crystallography Laboratory, Instituto de Tecnologia Química e Biológica, ITQB-NOVA, Oeiras, Portugal
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Fátima Serrano
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
| | - Helena Soares
- Human Immunobiology and Pathogenesis Group, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
- iNOVA4Health, Lisbon, Portugal
| |
Collapse
|
27
|
Han Q, Hu Y, Lu Z, Wang J, Chen H, Mo Z, Luo X, Li A, Dan X, Li Y. Study on the characterization of grouper (Epinephelus coioides) immunoglobulin T and its positive cells. FISH & SHELLFISH IMMUNOLOGY 2021; 118:102-110. [PMID: 34481975 DOI: 10.1016/j.fsi.2021.08.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 08/10/2021] [Accepted: 08/30/2021] [Indexed: 06/13/2023]
Abstract
Immunoglobulins (Igs) play a vital role in the adaptive immunity of gnathostomes. IgT, a particular Ig class in teleost fishes, receives much attention concerning the mucosal immunity. While, the characteristic and function of Epinephelus coioides IgT is still unknown. In our study, a polyclonal antibody was first prepared with grouper IgT heavy chain recombinant protein. IgT was revealed to be polymeric in serum and mucus. In normal groupers, IgT had high expression level in head kidney and spleen, while little amount in gills, thymus, gut and liver. The number of IgT-positive cells in different tissues was in line with their IgT expression. Furthermore, IgT could coat fractional bacteria in the mucus. In conclusion, this research revealed the protein characteristic, basal expression and bacterial coverage of grouper IgT. This is the first study to identify the characteristic of grouper IgT and demonstrate the capacity of coating microbes.
Collapse
Affiliation(s)
- Qing Han
- College of Marine Science, South China Agricultural University, Guangzhou, 510640, China
| | - Yingtong Hu
- College of Marine Science, South China Agricultural University, Guangzhou, 510640, China
| | - Zijun Lu
- College of Marine Science, South China Agricultural University, Guangzhou, 510640, China
| | - Jiule Wang
- College of Marine Science, South China Agricultural University, Guangzhou, 510640, China
| | - Hongping Chen
- College of Marine Science, South China Agricultural University, Guangzhou, 510640, China
| | - Zequan Mo
- College of Marine Science, South China Agricultural University, Guangzhou, 510640, China
| | - Xiaochun Luo
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Anxing Li
- State Key Laboratory of Biocontrol/Guangdong Provincial Key Lab for Aquatic Economic Animals, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, Guangdong Province, China
| | - Xueming Dan
- College of Marine Science, South China Agricultural University, Guangzhou, 510640, China.
| | - Yanwei Li
- College of Marine Science, South China Agricultural University, Guangzhou, 510640, China.
| |
Collapse
|
28
|
Trincado V, Gala RP, Morales JO. Buccal and Sublingual Vaccines: A Review on Oral Mucosal Immunization and Delivery Systems. Vaccines (Basel) 2021; 9:vaccines9101177. [PMID: 34696284 PMCID: PMC8539688 DOI: 10.3390/vaccines9101177] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, most vaccines available on the market are for parental use; however, this may not be the best option on several occasions. Mucosal routes of administration such as intranasal, sublingual, and buccal generate great interest due to the benefits they offer. These range from increasing patient compliance to inducing a more effective immune response than that achieved through conventional routes. Due to the activation of the common mucosal immune system, it is possible to generate an effective systemic and local immune response, which is not achieved through parenteral administration. Protection against pathogens that use mucosal entry routes is provided by an effective induction of mucosal immunity. Mucosal delivery systems are being developed, such as films and microneedles, which have proven to be effective, safe, and easy to administer. These systems have multiple advantages over commonly used injections, which are simple to manufacture, stable at room temperature, painless for the patient since they do not require puncture. Therefore, these delivery systems do not require to be administered by medical personnel; in fact, they could be self-administered.
Collapse
Affiliation(s)
- Valeria Trincado
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
| | - Rikhav P. Gala
- Biotechnology Division, Center Mid-Atlantic, Fraunhofer USA, Newark, DE 19702, USA;
| | - Javier O. Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
- Correspondence:
| |
Collapse
|
29
|
Pathogenesis of IgA Nephropathy: Current Understanding and Implications for Development of Disease-Specific Treatment. J Clin Med 2021; 10:jcm10194501. [PMID: 34640530 PMCID: PMC8509647 DOI: 10.3390/jcm10194501] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022] Open
Abstract
IgA nephropathy, initially described in 1968 as a kidney disease with glomerular “intercapillary deposits of IgA-IgG”, has no disease-specific treatment and is a common cause of kidney failure. Clinical observations and laboratory analyses suggest that IgA nephropathy is an autoimmune disease wherein the kidneys are damaged as innocent bystanders due to deposition of IgA1-IgG immune complexes from the circulation. A multi-hit hypothesis for the pathogenesis of IgA nephropathy describes four sequential steps in disease development. Specifically, patients with IgA nephropathy have elevated circulating levels of IgA1 with some O-glycans deficient in galactose (galactose-deficient IgA1) and these IgA1 glycoforms are recognized as autoantigens by unique IgG autoantibodies, resulting in formation of circulating immune complexes, some of which deposit in glomeruli and activate mesangial cells to induce kidney injury. This proposed mechanism is supported by observations that (i) glomerular immunodeposits in patients with IgA nephropathy are enriched for galactose-deficient IgA1 glycoforms and the corresponding IgG autoantibodies; (ii) circulatory levels of galactose-deficient IgA1 and IgG autoantibodies predict disease progression; and (iii) pathogenic potential of galactose-deficient IgA1 and IgG autoantibodies was demonstrated in vivo. Thus, a better understanding of the structure–function of these immunoglobulins as autoantibodies and autoantigens will enable development of disease-specific treatments.
Collapse
|
30
|
Sobia P, Archary D. Preventive HIV Vaccines-Leveraging on Lessons from the Past to Pave the Way Forward. Vaccines (Basel) 2021; 9:vaccines9091001. [PMID: 34579238 PMCID: PMC8472969 DOI: 10.3390/vaccines9091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/03/2021] [Indexed: 12/05/2022] Open
Abstract
Almost four decades on, since the 1980’s, with hundreds of HIV vaccine candidates tested in both non-human primates and humans, and several HIV vaccines trials later, an efficacious HIV vaccine continues to evade us. The enormous worldwide genetic diversity of HIV, combined with HIV’s inherent recombination and high mutation rates, has hampered the development of an effective vaccine. Despite the advent of antiretrovirals as pre-exposure prophylaxis and preventative treatment, which have shown to be effective, HIV infections continue to proliferate, highlighting the great need for a vaccine. Here, we provide a brief history for the HIV vaccine field, with the most recent disappointments and advancements. We also provide an update on current passive immunity trials, testing proof of the concept of the most clinically advanced broadly neutralizing monoclonal antibodies for HIV prevention. Finally, we include mucosal immunity, the importance of vaccine-elicited immune responses and the challenges thereof in the most vulnerable environment–the female genital tract and the rectal surfaces of the gastrointestinal tract for heterosexual and men who have sex with men transmissions, respectively.
Collapse
Affiliation(s)
- Parveen Sobia
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
| | - Derseree Archary
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Nelson Mandela School of Medicine, University of KwaZulu-Natal, Durban 4001, South Africa;
- Department of Medical Microbiology, University of KwaZulu-Natal, Durban 4001, South Africa
- Correspondence: ; Tel.: +27-(0)-31-655-0540
| |
Collapse
|
31
|
Baltierra-Uribe SL, Montañez-Barragán A, Romero-Ramírez H, Klimov-Kravtchenko K, Martínez-Pedro KI, Sánchez-Salguero E, Camorlinga-Ponce M, Torres J, Santos-Argumedo L. Colostrum IgA1 antibodies recognize antigens from Helicobacter pylori and prevent cytoskeletal changes in human epithelial cells. Eur J Immunol 2021; 51:2641-2650. [PMID: 34398472 DOI: 10.1002/eji.202049117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/11/2021] [Accepted: 08/13/2021] [Indexed: 11/06/2022]
Abstract
Helicobacter pylori is a gram-negative bacterium found on the luminal surface of the gastric mucosa in at least 50% of the world's human population. The protective effect of breastfeeding against H. pylori infection has been extensively reported; however, the mechanisms behind this protection remain poorly understood. Human IgA from colostrum has reactivity against H. pylori antigens. Despite that IgA1 and IgA2 display structural and functional differences, their reactivity against H. pylori had not been previously determined. We attested titers and reactivity of human colostrum-IgA subclasses by ELISA, immunoblot, and flow cytometry. Colostrum samples from healthy mothers had higher titers of IgA; and IgA1 mostly recognized H. pylori antigens. Moreover, we found a correlation between IgA1 reactivity and their neutralizing effect determined by inhibition of cytoskeletal changes in AGS cells infected with H. pylori. In conclusion, colostrum-IgA reduces H. pylori infection of epithelial gastric cells, suggesting an important role in preventing the bacteria establishment during the first months of life. As a whole, these results suggest that IgA1 from human colostrum provides protection that may help in the development of the mucosal immune system of newborn children. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shantal Lizbeth Baltierra-Uribe
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Alejandra Montañez-Barragán
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Héctor Romero-Ramírez
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Ksenia Klimov-Kravtchenko
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico.,University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan, Jalisco, Mexico
| | - Karla Ivette Martínez-Pedro
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico.,"La Cañada" University (UNCA), Oaxaca, Mexico
| | - Erick Sánchez-Salguero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Margarita Camorlinga-Ponce
- Infectious and Parasitic Disease Medical Research Unit (UIMEIP) at Pediatric Hospital in National Medical Center (CMN- SIGLO XXI) Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Javier Torres
- Infectious and Parasitic Disease Medical Research Unit (UIMEIP) at Pediatric Hospital in National Medical Center (CMN- SIGLO XXI) Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Leopoldo Santos-Argumedo
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
32
|
Ohyama Y, Renfrow MB, Novak J, Takahashi K. Aberrantly Glycosylated IgA1 in IgA Nephropathy: What We Know and What We Don't Know. J Clin Med 2021; 10:jcm10163467. [PMID: 34441764 PMCID: PMC8396900 DOI: 10.3390/jcm10163467] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/26/2021] [Accepted: 08/02/2021] [Indexed: 12/17/2022] Open
Abstract
IgA nephropathy (IgAN), the most common primary glomerular disease worldwide, is characterized by glomerular deposition of IgA1-containing immune complexes. The IgA1 hinge region (HR) has up to six clustered O-glycans consisting of Ser/Thr-linked N-acetylgalactosamine usually with β1,3-linked galactose and variable sialylation. Circulating levels of IgA1 with abnormally O-glycosylated HR, termed galactose-deficient IgA1 (Gd-IgA1), are increased in patients with IgAN. Current evidence suggests that IgAN is induced by multiple sequential pathogenic steps, and production of aberrantly glycosylated IgA1 is considered the initial step. Thus, the mechanisms of biosynthesis of aberrantly glycosylated IgA1 and the involvement of aberrant glycoforms of IgA1 in disease development have been studied. Furthermore, Gd-IgA1 represents an attractive biomarker for IgAN, and its clinical significance is still being evaluated. To elucidate the pathogenesis of IgAN, it is important to deconvolute the biosynthetic origins of Gd-IgA1 and characterize the pathogenic IgA1 HR O-glycoform(s), including the glycan structures and their sites of attachment. These efforts will likely lead to development of new biomarkers. Here, we review the IgA1 HR O-glycosylation in general and the role of aberrantly glycosylated IgA1 in the pathogenesis of IgAN in particular.
Collapse
Affiliation(s)
- Yukako Ohyama
- Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan;
| | - Matthew B. Renfrow
- Departments of Biochemistry and Molecular Genetics and Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.B.R.); (J.N.)
| | - Jan Novak
- Departments of Biochemistry and Molecular Genetics and Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (M.B.R.); (J.N.)
| | - Kazuo Takahashi
- Department of Biomedical Molecular Sciences, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan;
- Correspondence: ; Tel.: +81-(562)-93-2430; Fax: +81-(562)-93-1830
| |
Collapse
|
33
|
Meihua M, Xiaozhong L, Qin W, Yunfen Z, Yanyan C, Xunjun S. Association between Tfh and PGA in children with Henoch-Schönlein purpura. Open Med (Wars) 2021; 16:986-991. [PMID: 34250253 PMCID: PMC8254571 DOI: 10.1515/med-2021-0318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 05/25/2021] [Accepted: 06/07/2021] [Indexed: 11/15/2022] Open
Abstract
Objective The aim of this study was to investigate the roles of follicular helper CD4+ T cells (Tfh) and serum anti-α-1,4-d-polygalacturonic acid (PGA) antibody in the pathogenesis of Henoch-Schönlein purpura (HSP). Methods ELISA was performed to determine serum PGA-IgA and PGA-IgG. Flow cytometry was utilized to determine the peripheral CD4+ CXCR5+ and CD4+ CXCR5+ ICOS+ Tfh cells. Real-time PCR was conducted to determine the expression of Bcl-6 gene. Then the change of Tfh cells was analyzed, together with the association with the anti-PGA antibody as well as the roles in the pathogenesis of HSP. Results Compared with the cases with acute respiratory infection and elective surgery, the proportion of CD4+ CXCR5+ and CD4+ CXCR5+ ICOS+ Tfh cells in the HSP group showed significant elevation (P < 0.001). A significant correlation was noticed between PGA-IgA and CD4+ CXCR5+ Tfh cells (r = 0.380 and P = 0.042) and CD4+ CXCR5+ ICOS+ Tfh cells (r = 0.906 and P < 0.001). The expression of Bcl-6 in the HSP group showed no statistical difference compared with that in the acute respiratory infection and the surgery control (P < 0.05). Conclusion Increased activity of Tfh cells, which is closely related to mucosal immunity, may be a major contributor in the elevation of PGA-IgA, and Tfh cells and PGA-IgA are closely related to the occurrence of HSP.
Collapse
Affiliation(s)
- Miao Meihua
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Li Xiaozhong
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Wang Qin
- Department of Immunology, School of Biology & Basic Medical Science, Soochow University, Suzhou 215021, China
| | - Zhu Yunfen
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Cui Yanyan
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Shao Xunjun
- Department of Clinical Laboratory, Children's Hospital of Soochow University, Suzhou 215025, China
| |
Collapse
|
34
|
Ferreira SCM, Veiga MM, Hofer H, East ML, Czirják GÁ. Noninvasively measured immune responses reflect current parasite infections in a wild carnivore and are linked to longevity. Ecol Evol 2021; 11:7685-7699. [PMID: 34188844 PMCID: PMC8216923 DOI: 10.1002/ece3.7602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 01/03/2023] Open
Abstract
Host immune defenses are important components of host-parasite interactions that affect the outcome of infection and may have fitness consequences for hosts when increased allocation of resources to immune responses undermines other essential life processes. Research on host-parasite interactions in large free-ranging wild mammals is currently hampered by a lack of verified noninvasive assays. We successfully adapted existing assays to measure innate and adaptive immune responses produced by the gastrointestinal mucosa in spotted hyena (Crocuta crocuta) feces, including enzyme-linked immunosorbent assays (ELISAs), to quantify fecal immunoglobulins (total IgA, total IgG) and total fecal O-linked oligosaccharides (mucin). We investigated the effect of infection load by an energetically costly hookworm (Ancylostoma), parasite richness, host age, sex, year of sampling, and clan membership on immune responses and asked whether high investment in immune responses during early life affects longevity in individually known spotted hyenas in the Serengeti National Park, Tanzania. Fecal concentrations of IgA, IgG, and mucin increased with Ancylostoma egg load and were higher in juveniles than in adults. Females had higher mucin concentrations than males. Juvenile females had higher IgG concentrations than juvenile males, whereas adult females had lower IgG concentrations than adult males. High IgA concentrations during the first year of life were linked to reduced longevity after controlling for age at sampling and Ancylostoma egg load. Our study demonstrates that the use of noninvasive methods can increase knowledge on the complex relationship between gastrointestinal parasites and host local immune responses in wild large mammals and reveal fitness-relevant effects of these responses.
Collapse
Affiliation(s)
- Susana C. M. Ferreira
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
- Present address:
Division of Computational Systems BiologyCentre for Microbiology and Environmental Systems ScienceViennaAustria
| | - Miguel M. Veiga
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
| | - Heribert Hofer
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
- Department of Veterinary MedicineFreie Universität BerlinBerlinGermany
- Department of Biology, Chemistry and PharmacyFreie Universität BerlinBerlinGermany
| | - Marion L. East
- Department of Ecological DynamicsLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
| | - Gábor Á. Czirják
- Department of Wildlife DiseasesLeibniz Institute for Zoo and Wildlife ResearchBerlinGermany
| |
Collapse
|
35
|
Classification, structural biology, and applications of mucin domain-targeting proteases. Biochem J 2021; 478:1585-1603. [DOI: 10.1042/bcj20200607] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022]
Abstract
Epithelial surfaces throughout the body are coated by mucins, a class of proteins carrying domains characterized by a high density of O-glycosylated serine and threonine residues. The resulting mucosal layers form crucial host-microbe interfaces that prevent the translocation of microbes while also selecting for distinct bacteria via the presented glycan repertoire. The intricate interplay between mucus production and breakdown thus determines the composition of the microbiota maintained within these mucosal environments, which can have a large influence on the host during both homeostasis and disease. Most research to date on mucus breakdown has focused on glycosidases that trim glycan structures to release monosaccharides as a source of nutrients. More recent work has uncovered the existence of mucin-type O-glycosylation-dependent proteases that are secreted by pathogens, commensals, and mutualists to facilitate mucosal colonization and penetration. Additionally, immunoglobulin A (IgA) proteases promote bacterial colonization in the presence of neutralizing secretory IgA through selective cleavage of the heavily O-glycosylated hinge region. In this review, we summarize families of O-glycoproteases and IgA proteases, discuss known structural features, and review applications of these enzymes to glycobiology.
Collapse
|
36
|
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
|
37
|
Seaton KE, Deal A, Han X, Li SS, Clayton A, Heptinstall J, Duerr A, Allen MA, Shen X, Sawant S, Yates NL, Spearman P, Churchyard G, Goepfert PA, Maenza J, Gray G, Pantaleo G, Polakowski L, Robinson HL, Grant S, Randhawa AK, Huang Y, Morgan C, Grunenberg N, Karuna S, Gilbert PB, McElrath MJ, Huang Y, Tomaras GD. Meta-analysis of HIV-1 vaccine elicited mucosal antibodies in humans. NPJ Vaccines 2021; 6:56. [PMID: 33859204 PMCID: PMC8050318 DOI: 10.1038/s41541-021-00305-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 02/24/2021] [Indexed: 12/22/2022] Open
Abstract
We studied mucosal immune responses in six HIV-1 vaccine trials investigating different envelope (Env)-containing immunogens. Regimens were classified into four categories: DNA/vector, DNA/vector plus protein, protein alone, and vector alone. We measured HIV-1-specific IgG and IgA in secretions from cervical (n = 111) and rectal swabs (n = 154), saliva (n = 141), and seminal plasma (n = 124) and compared to corresponding blood levels. Protein-containing regimens had up to 100% response rates and the highest Env-specific IgG response rates. DNA/vector groups elicited mucosal Env-specific IgG response rates of up to 67% that varied across specimen types. Little to no mucosal IgA responses were observed. Overall, gp41- and gp140-specific antibodies dominated gp120 mucosal responses. In one trial, prior vaccination with a protein-containing immunogen maintained durability of cervical and rectal IgG for up to 17 years. Mucosal IgG responses were boosted after revaccination. These findings highlight a role for protein immunization in eliciting HIV-1-specific mucosal antibodies and the ability of HIV-1 vaccines to elicit durable HIV-1-specific mucosal IgG.
Collapse
Affiliation(s)
- Kelly E Seaton
- Duke Human Vaccine Institute, Durham, NC, USA.
- Department of Surgery, Duke University, Durham, NC, USA.
- Department of Immunology, Duke University, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA.
| | - Aaron Deal
- Duke Human Vaccine Institute, Durham, NC, USA
| | - Xue Han
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shuying S Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ashley Clayton
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jack Heptinstall
- Duke Human Vaccine Institute, Durham, NC, USA
- Department of Surgery, Duke University, Durham, NC, USA
| | - Ann Duerr
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | - Sheetal Sawant
- Duke Human Vaccine Institute, Durham, NC, USA
- Department of Surgery, Duke University, Durham, NC, USA
| | - Nicole L Yates
- Duke Human Vaccine Institute, Durham, NC, USA
- Department of Surgery, Duke University, Durham, NC, USA
| | - Paul Spearman
- Division of Infectious Diseases, Cincinnati Children's Hospital, Cincinnatti, OH, USA
| | - Gavin Churchyard
- Aurum Institute, Johannesburg, South Africa
- School of Public Health, University of Witwatersrand, Johannesburg, South Africa
| | - Paul A Goepfert
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Janine Maenza
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Glenda Gray
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- South African Medical Research Council, Cape Town, South Africa
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, and Swiss Vaccine Research Institute, Lausanne University Hospital, Lausanne, Switzerland
| | | | | | - Shannon Grant
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - April K Randhawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Cecilia Morgan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicole Grunenberg
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shelly Karuna
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Georgia D Tomaras
- Duke Human Vaccine Institute, Durham, NC, USA.
- Department of Surgery, Duke University, Durham, NC, USA.
- Department of Immunology, Duke University, Durham, NC, USA.
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA.
| |
Collapse
|
38
|
Almaghlouth I, Johnson SR, Pullenayegum E, Gladman D, Urowitz M. Immunoglobulin levels in systemic lupus erythematosus: A narrative review. Lupus 2021; 30:867-875. [PMID: 33779380 DOI: 10.1177/09612033211004714] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Immunoglobulins play a fundamental role in the protection of the human body against internal and external threats. They also contribute to the immune system homeostasis and maintenance of self-tolerance. Hypogammaglobulinemia is occasionally encountered in routine clinical practice by rheumatologists. Low levels of immunoglobulins can occur as primary or secondary issues and may predispose patients to various forms of infection. However, the impact of the low immunoglobulin level abnormality varies with the underlying condition. In this narrative review, we shed light on the overall types and functions of immunoglobulins for clinicians. We discuss important principles of immunoglobulin measurements. We then consider the primary and secondary causes of low immunoglobulins with a special focus on hypogammaglobulinemia in patients with systemic lupus erythematosus (SLE).
Collapse
Affiliation(s)
- Ibrahim Almaghlouth
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, Mount Sinai Hospital, University of Toronto and the Schroeder Arthritis Institute, University Health Network, Toronto, Canada.,Rheumatology Unit, Department of Medicine, King Saud University, Riyadh, Saudi Arabia.,College of Medicine Research Center, King Saud University, Riyadh, Saudi Arabia
| | - Sindhu R Johnson
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, Mount Sinai Hospital, University of Toronto and the Schroeder Arthritis Institute, University Health Network, Toronto, Canada.,Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada
| | - Eleanor Pullenayegum
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada.,Program in Child Health Evaluative Sciences, SickKids Research Institute, Toronto, Canada
| | - Dafna Gladman
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, Mount Sinai Hospital, University of Toronto and the Schroeder Arthritis Institute, University Health Network, Toronto, Canada.,Centre for Prognosis in Rheumatic Diseases, University Health Network, Toronto, Canada
| | - Murray Urowitz
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, Mount Sinai Hospital, University of Toronto and the Schroeder Arthritis Institute, University Health Network, Toronto, Canada.,Centre for Prognosis in Rheumatic Diseases, University Health Network, Toronto, Canada
| |
Collapse
|
39
|
Influences of Selenium-Enriched Yeast on Growth Performance, Immune Function, and Antioxidant Capacity in Weaned Pigs Exposure to Oxidative Stress. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5533210. [PMID: 33855070 PMCID: PMC8019624 DOI: 10.1155/2021/5533210] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/26/2021] [Accepted: 03/18/2021] [Indexed: 02/07/2023]
Abstract
This study elucidated the function role of dietary selenium-enriched yeast (SeY) supplementation on growth performance, immune function, and antioxidant capacity in weaned pigs exposure to oxidative stress. Thirty-two similarity weight pigs were randomly divided into four treatments: (1) nonchallenged control, (2) control+SeY, (3) control+diquat, and (4) control+SeY+diquat. The period of experiment was 21 days; on day 16, pigs were injected with diquat or sterile saline. Results revealed that oxidative stress was notably detrimental to the growth performance of piglets, but SeY supplementation ameliorated this phenomenon, which might be regarding the increasing of body antioxidant capacity and immune functions. In details, SeY supplementation improved the digestibility of crude protein (CP), ash, and gross energy (GE). Moreover, the serum concentrations of proinflammatory cytokines (TNF-α, IL-1β, and IL-6), glutamic-pyruvic transaminase(GPT), and glutamic-oxaloacetic transaminase (GOT) were reduced via SeY supplemented, and serum concentrations of immunoglobulins A (IgA), IgG, and activities of antioxidant enzymes such as the superoxide dismutase (SOD), catalase (CAT) ,and glutathione peroxidase (GSH-Px) were improved in the diquat-challenged pigs (P < 0.05). In addition, SeY supplementation acutely enhanced the activities of these antioxidant enzymes in the liver and thymus upon diquat challenge, which involved with the upregulation of the critical genes related antioxidant signaling such as the nuclear factor erythroid-derived 2-related factor 2 (Nrf-2) and heme oxygenase-1 (HO-1) (P < 0.05). Importantly, we also found that SeY supplementation apparently reduced the malondialdehyde (MDA) concentrations in the liver, thymus, and serum (P < 0.05). Specifically, the expression levels of TNF-α, IL-6, IL-1β, Toll-like receptor 4 (TLR-4), and nuclear factor-κB (NF-κB) in the liver and thymus were downregulated by SeY upon diquat challenge. These results suggested that SeY can attenuate oxidative stress-induced growth retardation, which was associated with elevating body antioxidant capacity, immune functions, and suppressed inflammatory response.
Collapse
|
40
|
Chen M, Zhao Q, Diao L, Xue K, Ruan Y, Xue F, Li J, Shi R, Pan M, Zheng J, Cao H. Distribution of anti-melanoma differentiation associated gene 5 (MDA5) IgG subclasses in MDA5+ dermatomyositis. Rheumatology (Oxford) 2021; 61:430-439. [PMID: 33742662 DOI: 10.1093/rheumatology/keab268] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES The anti-melanoma differentiation-associated gene 5 (MDA5) antibody is the main predictor of interstitial lung disease (ILD) in dermatomyositis (DM) and clinically amyopathic dermatomyositis (CADM). Nevertheless, a subset of MDA5+ patients have a favorable prognosis. We aimed to determine the possibility of using anti-MDA5 antibody isotypes and IgG subclasses for evaluating ILD risk. METHODS The isotypes (IgG, IgA and IgM) of anti-MDA5 were detected in serum samples of 36 anti-MDA5+ patients with DM/CADM using enzyme-linked immunosorbent assay (ELISA). IgG subclasses of anti-MDA5 antibodies were further investigated. Laboratory findings and cumulative survival were analyzed based on the isotypes of anti-MDA5 and subclasses of anti-MDA5 IgG. RESULTS Among the MDA5+ patients with DM/CADM, the positive rates of anti-MDA5 IgG, IgA, IgM were 100%, 97%, and 6%, respectively. The positive rates of anti-MDA5 IgG1, IgG2, IgG3, and IgG4 were 72%, 25%, 0%, and 28%, respectively. The incidence of acute interstitial pneumonia, mortality rate, and serum ferritin were significantly higher in anti-MDA5 IgG1+ patients than those in anti-MDA5 IgG1- patients with DM/CADM (P = 0.0027, 0.015, 0.0011, respectively). The sensitivity and specificity of anti-MDA5 IgG1 for predicting mortality were 100% and 41.7%, respectively. A combination of anti-MDA5 IgG1 and IgG4 for predicting mortality, yielded better specificity (87.5%). CONCLUSION IgA and IgG are the primary anti-MDA5 antibody isotypes. Anti-MDA5 IgG1 is the primary component of MDA5 IgG subclasses and anti-MDA5 IgG1 and IgG4 might serve as useful biomarkers for predicting mortality in DM-ILD.
Collapse
Affiliation(s)
- Mengya Chen
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Qian Zhao
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Licheng Diao
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Ke Xue
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Yeping Ruan
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Feng Xue
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Jian Li
- Department of Clinical Research Center, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Ruofei Shi
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Meng Pan
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Jie Zheng
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| | - Hua Cao
- Department of Dermatology, Rui Jin Hospital, School of Medicine, Shanghai Jiao Tong University, 200025 Shanghai, China
| |
Collapse
|
41
|
Onabajo OO, Mattapallil JJ. Gut Microbiome Homeostasis and the CD4 T- Follicular Helper Cell IgA Axis in Human Immunodeficiency Virus Infection. Front Immunol 2021; 12:657679. [PMID: 33815419 PMCID: PMC8017181 DOI: 10.3389/fimmu.2021.657679] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
Human Immunodeficiency Virus (HIV) and Simian Immunodeficiency Virus (SIV) are associated with severe perturbations in the gut mucosal environment characterized by massive viral replication and depletion of CD4 T cells leading to dysbiosis, breakdown of the epithelial barrier, microbial translocation, immune activation and disease progression. Multiple mechanisms play a role in maintaining homeostasis in the gut mucosa and protecting the integrity of the epithelial barrier. Among these are the secretory IgA (sIgA) that are produced daily in vast quantities throughout the mucosa and play a pivotal role in preventing commensal microbes from breaching the epithelial barrier. These microbe specific, high affinity IgA are produced by IgA+ plasma cells that are present within the Peyer’s Patches, mesenteric lymph nodes and the isolated lymphoid follicles that are prevalent in the lamina propria of the gastrointestinal tract (GIT). Differentiation, maturation and class switching to IgA producing plasma cells requires help from T follicular helper (Tfh) cells that are present within these lymphoid tissues. HIV replication and CD4 T cell depletion is accompanied by severe dysregulation of Tfh cell responses that compromises the generation of mucosal IgA that in turn alters barrier integrity leading to commensal bacteria readily breaching the epithelial barrier and causing mucosal pathology. Here we review the effect of HIV infection on Tfh cells and mucosal IgA responses in the GIT and the consequences these have for gut dysbiosis and mucosal immunopathogenesis.
Collapse
Affiliation(s)
- Olusegun O Onabajo
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Joseph J Mattapallil
- F. E. Hebert School of Medicine, Uniformed Services University, Bethesda, MD, United States
| |
Collapse
|
42
|
Functional Role of B Cells in Atherosclerosis. Cells 2021; 10:cells10020270. [PMID: 33572939 PMCID: PMC7911276 DOI: 10.3390/cells10020270] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/30/2022] Open
Abstract
Atherosclerosis is a lipid-driven inflammatory disease of blood vessels, and both innate and adaptive immune responses are involved in its development. The impact of B cells on atherosclerosis has been demonstrated in numerous studies and B cells have been found in close proximity to atherosclerotic plaques in humans and mice. B cells exert both atheroprotective and pro-atherogenic functions, which have been associated with their B cell subset attribution. While B1 cells and marginal zone B cells are considered to protect against atherosclerosis, follicular B cells and innate response activator B cells have been shown to promote atherosclerosis. In this review, we shed light on the role of B cells from a different, functional perspective and focus on the three major B cell functions: antibody production, antigen presentation/T cell interaction, and the release of cytokines. All of these functions have the potential to affect atherosclerosis by multiple ways and are dependent on the cellular milieu and the activation status of the B cell. Moreover, we discuss B cell receptor signaling and the mechanism of B cell activation under atherosclerosis-prone conditions. By summarizing current knowledge of B cells in and beyond atherosclerosis, we are pointing out open questions and enabling new perspectives.
Collapse
|
43
|
Pascal V, Hiblot M, Wehbi B, Aldigier JC, Cogné M. [Microbiota and IgA response homeostasis]. Med Sci (Paris) 2021; 37:35-40. [PMID: 33492216 DOI: 10.1051/medsci/2020258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mucosal immunity has to deal with a patchy mix of commensal but also eventually pathogenic bugs. Immunoglobulins of the A class (IgA) are opposing to this duality a functional balance going from tolerance to protective response or even to hyper-inflammation. Recent reports have shown the binding of polyreactive natural IgA, but also of affinity maturated protective IgA to the commensal microbiota, to superantigens and also to vaccinal antigens. Diverse types of humoral responses thus altogether contribute to the homeostasis of mucosal immunity. Their knowledge has to be taken into consideration for defining strategies of immuno-intervention, for mucosal vaccination as much as for immunotherapy of chronic inflammatory bowel disease.
Collapse
Affiliation(s)
- Virginie Pascal
- CNRS UMR 7276, Inserm U1262, Contrôle de la réponse immune B et lymphoproliférations, Université de Limoges, Rue du Docteur Marcland, 87000 Limoges, France
| | - Margaux Hiblot
- CNRS UMR 7276, Inserm U1262, Contrôle de la réponse immune B et lymphoproliférations, Université de Limoges, Rue du Docteur Marcland, 87000 Limoges, France
| | - Batoul Wehbi
- CNRS UMR 7276, Inserm U1262, Contrôle de la réponse immune B et lymphoproliférations, Université de Limoges, Rue du Docteur Marcland, 87000 Limoges, France
| | - Jean-Claude Aldigier
- CNRS UMR 7276, Inserm U1262, Contrôle de la réponse immune B et lymphoproliférations, Université de Limoges, Rue du Docteur Marcland, 87000 Limoges, France
| | - Michel Cogné
- CNRS UMR 7276, Inserm U1262, Contrôle de la réponse immune B et lymphoproliférations, Université de Limoges, Rue du Docteur Marcland, 87000 Limoges, France - EFS Bretagne, Inserm U1236, Université de Rennes 1, Rue Pierre-Jean-Gineste, 35000 Rennes, France
| |
Collapse
|
44
|
Galipeau Y, Greig M, Liu G, Driedger M, Langlois MA. Humoral Responses and Serological Assays in SARS-CoV-2 Infections. Front Immunol 2020; 11:610688. [PMID: 33391281 PMCID: PMC7775512 DOI: 10.3389/fimmu.2020.610688] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022] Open
Abstract
In December 2019, the novel betacoronavirus Severe Acute Respiratory Disease Coronavirus 2 (SARS-CoV-2) was first detected in Wuhan, China. SARS-CoV-2 has since become a pandemic virus resulting in hundreds of thousands of deaths and deep socioeconomic implications worldwide. In recent months, efforts have been directed towards detecting, tracking, and better understanding human humoral responses to SARS-CoV-2 infection. It has become critical to develop robust and reliable serological assays to characterize the abundance, neutralization efficiency, and duration of antibodies in virus-exposed individuals. Here we review the latest knowledge on humoral immune responses to SARS-CoV-2 infection, along with the benefits and limitations of currently available commercial and laboratory-based serological assays. We also highlight important serological considerations, such as antibody expression levels, stability and neutralization dynamics, as well as cross-reactivity and possible immunological back-boosting by seasonal coronaviruses. The ability to accurately detect, measure and characterize the various antibodies specific to SARS-CoV-2 is necessary for vaccine development, manage risk and exposure for healthcare and at-risk workers, and for monitoring reinfections with genetic variants and new strains of the virus. Having a thorough understanding of the benefits and cautions of standardized serological testing at a community level remains critically important in the design and implementation of future vaccination campaigns, epidemiological models of immunity, and public health measures that rely heavily on up-to-date knowledge of transmission dynamics.
Collapse
Affiliation(s)
- Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew Greig
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - George Liu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | | | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
- uOttawa Center for Infection, Immunity and Inflammation (CI3), Ottawa, ON, Canada
| |
Collapse
|
45
|
Xu Z, Takizawa F, Casadei E, Shibasaki Y, Ding Y, Sauters TJC, Yu Y, Salinas I, Sunyer JO. Specialization of mucosal immunoglobulins in pathogen control and microbiota homeostasis occurred early in vertebrate evolution. Sci Immunol 2020; 5:5/44/eaay3254. [PMID: 32034088 DOI: 10.1126/sciimmunol.aay3254] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/16/2020] [Indexed: 12/11/2022]
Abstract
Although mammalian secretory immunoglobulin A (sIgA) targets mucosal pathogens for elimination, its interaction with the microbiota also enables commensal colonization and homeostasis. This paradoxical requirement in the control of pathogens versus microbiota raised the question of whether mucosal (secretory) Igs (sIgs) evolved primarily to protect mucosal surfaces from pathogens or to maintain microbiome homeostasis. To address this central question, we used a primitive vertebrate species (rainbow trout) in which we temporarily depleted its mucosal Ig (sIgT). Fish devoid of sIgT became highly susceptible to a mucosal parasite and failed to develop compensatory IgM responses against it. IgT depletion also induced a profound dysbiosis marked by the loss of sIgT-coated beneficial taxa, expansion of pathobionts, tissue damage, and inflammation. Restitution of sIgT levels in IgT-depleted fish led to a reversal of microbial translocation and tissue damage, as well as to restoration of microbiome homeostasis. Our findings indicate that specialization of sIgs in pathogen and microbiota control occurred concurrently early in evolution, thus revealing primordially conserved principles under which primitive and modern sIgs operate in the control of microbes at mucosal surfaces.
Collapse
Affiliation(s)
- Zhen Xu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Fumio Takizawa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.,Faculty of Marine Science and Technology, Fukui Prefectural University, Obama, Fukui 917-0003, Japan
| | - Elisa Casadei
- Center for Evolutionary and Theoretical Immunology (CETI), Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Yasuhiro Shibasaki
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yang Ding
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Thomas J C Sauters
- Center for Evolutionary and Theoretical Immunology (CETI), Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA
| | - Yongyao Yu
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Irene Salinas
- Center for Evolutionary and Theoretical Immunology (CETI), Department of Biology, University of New Mexico, Albuquerque, NM 87131, USA.
| | - J Oriol Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
46
|
Ejemel M, Gawron MA, Schneider MI, Wallace A, Schiller ZA, Schneider R, Martin III JC, Klempner MS, Wang Y, Cavacini LA. Highly Specific Mouse Anti-Joining Chain of Human Immunoglobulin A. Monoclon Antib Immunodiagn Immunother 2020; 39:228-232. [DOI: 10.1089/mab.2020.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Monir Ejemel
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Melissa A. Gawron
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Matthew I. Schneider
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Aaron Wallace
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Zachary A. Schiller
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Ryan Schneider
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Joseph C. Martin III
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Mark S. Klempner
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Yang Wang
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| | - Lisa A. Cavacini
- MassBiologics, University of Massachusetts Medical School, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Scheepers C, Bekker V, Anthony C, Richardson SI, Oosthuysen B, Moyo T, Kgagudi P, Kitchin D, Nonyane M, York T, Mielke D, Mabvakure BM, Sheng Z, Lambson BE, Ismail A, Garrett NJ, Abdool Karim SS, Shapiro L, Williamson C, Morris L, Moore PL. Antibody Isotype Switching as a Mechanism to Counter HIV Neutralization Escape. Cell Rep 2020; 33:108430. [PMID: 33238131 PMCID: PMC7723817 DOI: 10.1016/j.celrep.2020.108430] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/18/2020] [Accepted: 11/02/2020] [Indexed: 02/04/2023] Open
Abstract
Neutralizing antibodies (nAbs) to highly variable viral pathogens show remarkable diversification during infection, resulting in an “arms race” between virus and host. Studies of nAb lineages have shown how somatic hypermutation (SHM) in immunoglobulin (Ig)-variable regions enables maturing antibodies to neutralize emerging viral escape variants. However, the Ig-constant region (which determines isotype) can also influence epitope recognition. Here, we use longitudinal deep sequencing of an HIV-directed nAb lineage, CAP88-CH06, and identify several co-circulating isotypes (IgG3, IgG1, IgA1, IgG2, and IgA2), some of which share identical variable regions. First, we show that IgG3 and IgA1 isotypes are better able to neutralize longitudinal autologous viruses and epitope mutants than can IgG1. Second, detrimental class-switch recombination (CSR) events that resulted in reduced neutralization can be rescued by further CSR, which we term “switch redemption.” Thus, CSR represents an additional immunological mechanism to counter viral escape from HIV-specific antibody responses. Scheepers et al. show within an HIV-specific antibody lineage that isotypes confer variable ability to neutralize emerging viral escape variants. This suggests that class switching, in addition to somatic hypermutation of immunoglobulin-variable regions, contributes to antibody maturation during infection.
Collapse
Affiliation(s)
- Cathrine Scheepers
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Valerie Bekker
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Colin Anthony
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7701, South Africa
| | - Simone I Richardson
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Brent Oosthuysen
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Thandeka Moyo
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Prudence Kgagudi
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Dale Kitchin
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Molati Nonyane
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Talita York
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7701, South Africa
| | - Dieter Mielke
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7701, South Africa
| | - Batsirai M Mabvakure
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Zizhang Sheng
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA; Zuckerman Brain Mind Behaviour Institute, Columbia University, New York, NY 10027, USA
| | - Bronwen E Lambson
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa
| | - Arshad Ismail
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa
| | - Nigel J Garrett
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), KwaZulu-Natal 4013, South Africa
| | - Salim S Abdool Karim
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), KwaZulu-Natal 4013, South Africa; Department of Epidemiology, Columbia University, New York, NY 10032, USA
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA; Zuckerman Brain Mind Behaviour Institute, Columbia University, New York, NY 10027, USA
| | - Carolyn Williamson
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7701, South Africa
| | - Lynn Morris
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa; Centre for the AIDS Programme of Research in South Africa (CAPRISA), KwaZulu-Natal 4013, South Africa.
| | - Penny L Moore
- Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2131, South Africa; Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg 2050, South Africa; Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7701, South Africa; Centre for the AIDS Programme of Research in South Africa (CAPRISA), KwaZulu-Natal 4013, South Africa.
| |
Collapse
|
48
|
Jones K, Savulescu AF, Brombacher F, Hadebe S. Immunoglobulin M in Health and Diseases: How Far Have We Come and What Next? Front Immunol 2020; 11:595535. [PMID: 33193450 PMCID: PMC7662119 DOI: 10.3389/fimmu.2020.595535] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
B lymphocytes are important in secreting antibodies that protect against invading pathogens such as viruses, bacteria, parasites, and also in mediating pathogenesis of allergic diseases and autoimmunity. B lymphocytes develop in the bone marrow and contain heavy and light chains, which upon ligation form an immunoglobulin M (IgM) B cell receptor (BCR) expressed on the surface of naïve immature B cells. Naïve B cells expressing either IgM or IgD isotypes are thought to play interchangeable functions in antibody responses to T cell-dependent and T cell-independent antigens. IgM short-lived plasma cells (SLPCs) and antigen-specific IgM memory B cells (MBCs-M) are critical in the first few days of infection, as well as long-term memory induced by vaccination, respectively. At mucosal surfaces, IgM is thought to play a critical part in promoting mucosal tolerance and shaping microbiota together with IgA. In this review, we explore how IgM structure and BCR signaling shapes B cell development, self and non-self-antigen-specific antibody responses, responses to infectious (such as viruses, parasites, and fungal) and non-communicable diseases (such as autoimmunity and allergic asthma). We also explore how metabolism could influence other B cell functions such as mucosal tolerance and class switching. Finally, we discuss some of the outstanding critical research questions in both experimental and clinical settings targeting IgM.
Collapse
Affiliation(s)
- Katelyn Jones
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Anca F. Savulescu
- Division of Chemical, Systems & Synthetic Biology, Faculty of Health Sciences, Institute of Infectious Disease & Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Frank Brombacher
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Immunology, Health Science Faculty, International Centre for Genetic Engineering and Biotechnology (ICGEB) and Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine (IDM), University of Cape Town, Cape Town, South Africa
| | - Sabelo Hadebe
- Division of Immunology, Department of Pathology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
49
|
Shao J, Yin Z, Wang Y, Yang Y, Tang Q, Zhang M, Jiao J, Liu C, Yang M, Zhen L, Hassouna A, White WL, Lu J. Effects of Different Doses of Eucalyptus Oil From Eucalyptus globulus Labill on Respiratory Tract Immunity and Immune Function in Healthy Rats. Front Pharmacol 2020; 11:1287. [PMID: 32973518 PMCID: PMC7472567 DOI: 10.3389/fphar.2020.01287] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Eucalyptol (1,8-cineole), the major constituent of eucalyptus oil (EO), was used in traditional medicine as a remedy for colds and bronchitis. This study aimed at clarifying the effect of eucalyptol on respiratory immune function of CD8 and CD4 cells, and alveolar macrophages (AM). Thirty male Sprague-Dawley rats were divided into experimental and control groups. The drug was given once a day for 3 weeks and the experimental group was divided according to the eucalyptol dose into: 30, 100, and 300 mg·kg-1 groups. Flow cytometry was used to detect the phagocytic function of CD4, CD8 cells, and AM in the bronchopulmonary lavage fluid. The 30 and 100 mg·kg-1 groups had an up-regulation effect on CD8 (p < 0.05), with no significant effect on macrophage phagocytosis. The 300 mg·kg-1 group had an inhibitory effect on CD8 and macrophage phagocytosis (p < 0.05), with no significant difference in CD4 between groups. Further investigation was conducted to evaluate the effect of EO on immune function in rats by detecting blood T, B, and NK cells using flow cytometry, and blood IgA, IgG, IgM, and IFN-γ levels by ELISA. High dosage of eucalyptol significantly reduced the proportion of blood B and NK cells (p < 0.05). IgA was decreased in the 100 and 300 mg·kg-1 groups (p < 0.05). There are no significant differences between the number of T cells and the IgG, IgM, and IFN-γ levels between experimental and control groups. Rational use of EO containing eucalyptol can improve the immune function of the respiratory tract and the body immunity, while high dose could have damaging effects, through modifying the phagocytic function of CD8 cells and reducing the proportion of blood B cells, NK cells, and IgA.
Collapse
Affiliation(s)
- Jie Shao
- 521 Hospital of Norinco Group, Xi'an, China
| | | | - Yaqin Wang
- 521 Hospital of Norinco Group, Xi'an, China
| | | | - Qing Tang
- 521 Hospital of Norinco Group, Xi'an, China
| | | | | | | | | | | | - Amira Hassouna
- School of Public Health and Interdisciplinary Studies, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand.,Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - William Lindsey White
- School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Jun Lu
- School of Public Health and Interdisciplinary Studies, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand.,School of Science, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand.,College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
50
|
Leya T, Ahmad I, Sharma R, Tripathi G, Kurcheti PP, Rajendran KV, Bedekar MK. Bicistronic DNA vaccine macromolecule complexed with poly lactic-co-glycolic acid-chitosan nanoparticles enhanced the mucosal immunity of Labeo rohita against Edwardsiella tarda infection. Int J Biol Macromol 2020; 156:928-937. [DOI: 10.1016/j.ijbiomac.2020.04.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/10/2020] [Accepted: 04/03/2020] [Indexed: 12/19/2022]
|