1
|
Du YZ, Liu JH, Zheng FC, Zhou Q, Guo J. The association between serum albumin-globulin ratio and overactive bladder in American adults: a cross-sectional study. Eur J Med Res 2025; 30:229. [PMID: 40176132 PMCID: PMC11963652 DOI: 10.1186/s40001-025-02441-6] [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: 07/05/2024] [Accepted: 03/08/2025] [Indexed: 04/04/2025] Open
Abstract
OBJECTIVES The albumin-globulin ratio (AGR) is considered an important indicator reflecting an individual's immune function and nutritional status, and it is closely associated with various health conditions. However, despite its widely studied correlations in numerous health fields, the link between AGR and Overactive Bladder (OAB) is still not completely comprehended. METHODS Data were sourced from the National Health and Nutrition Examination Survey (NHANES) database, selecting adult samples spanning from 2007 to 2018. Through comprehensive questionnaires and laboratory tests, we gathered data pertinent to OAB and the AGR. To explore the association between AGR levels and the likelihood of developing OAB, we utilized advanced statistical techniques, such as weighted multivariate logistic regression and restricted cubic spline (RCS) models. Furthermore, we carried out subgroup analyses to assess the uniformity of this association across various demographics. RESULTS After adjusting for relevant covariates, we discovered a marked negative correlation between AGR levels and the risk of OAB. As AGR increased, the incidence of OAB showed a declining trend (OR = 0.69; 95% CI 0.56-0.85). Furthermore, significant nonlinear dose-response relationship was observed between AGR levels and the risk of OAB (P < 0.001), and this association remained stable in stratified analyses. CONCLUSIONS Our results indicate that elevated AGR levels could be linked to a reduced risk of OAB. This observation highlights the potential role of AGR in assessing and preventing the occurrence of OAB.
Collapse
Affiliation(s)
- Yuan-Zhuo Du
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Nanchang, China
| | - Jia-Hao Liu
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Nanchang, China
| | - Fu-Chun Zheng
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Nanchang, China
| | - Qiang Zhou
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Nanchang, China.
| | - Ju Guo
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Key Laboratory of Urinary System Diseases of Jiangxi Province, Nanchang, China.
| |
Collapse
|
2
|
Davias A, Verghese M, Bridgman SL, Tun HM, Field CJ, Hicks M, Pei J, Hicks A, Moraes TJ, Simons E, Turvey SE, Subbarao P, Scott JA, Mandhane PJ, Kozyrskyj AL. Gut microbiota metabolites, secretory immunoglobulin A and Bayley-III cognitive scores in children from the CHILD Cohort Study. Brain Behav Immun Health 2025; 44:100946. [PMID: 39911944 PMCID: PMC11795817 DOI: 10.1016/j.bbih.2025.100946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/19/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
Background Dysbiosis of the gut microbiota has been demonstrated in neurodevelopmental disorders but the underlying mechanisms that may explain these associations are poorly understood. Gut secretory immunoglobulin A (SIgA) binds pathogenic microbes, preventing mucosal penetration. Gut microbes also influence SIgA production and its binding characteristics through short-chain fatty acid (SCFA) metabolites, allowing them to regulate the immune response. Serum IgA deficiency has been noted in children with autism spectrum disorders (ASD). In this study, we aimed to determine whether SIgA level in infancy is associated with gut microbiota taxonomy and metabolites, and neurodevelopmental outcomes in preschool children. Methods For a subsample of 178 children from the Canadian CHILD Cohort Study, gut microbiota of fecal samples collected at 3-4 months and 12 months was profiled using 16S rRNA sequencing. Gut bacterial metabolites levels and SIgA level were measured by nuclear magnetic resonance (NMR) based metabolomics and SIgA enzyme-linked immunosorbent assay at 3-4 months, respectively. Bayley-III Scale of Infant Development was assessed at 12 and 24 months. We evaluated direct relationships in multiple linear regression models and putative causal relationships in statistical mediation models. Results Propionate and butyrate levels at 3-4 months were associated with decreased Bayley cognitive score at 24 months (p-values: 0.01 and 0.02, respectively) in adjusted multiple linear regression models, but when we investigated an indirect relationship mediated by decreased SIgA level at 3-4 months, it did not reach statistical significance (p-values: 0.18 and 0.20, respectively). Lactate level at 3-4 months was associated with increased Bayley cognitive score at 24 months in adjusted multiple linear regression models (p-value: 0.01), but the statistical model mediated by increased SIgA level at 3-4 months did not reach statistical significance neither (p-value: 0.20). Conclusions Our study contributes to growing evidence that neurodevelopment is influenced by the infant gut microbiota and that it might involve SIgA level, but larger studies are required.
Collapse
Affiliation(s)
- Aline Davias
- Edmonton Clinic Health Academy, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Environmental Epidemiology Applied to Development and Respiratory Health Team, Institute for Advanced Biosciences, University Grenoble Alpes, Inserm, CNRS, 38700, La Tronche, France
| | - Myah Verghese
- Edmonton Clinic Health Academy, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Sarah L. Bridgman
- Edmonton Clinic Health Academy, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Hein M. Tun
- The Jockey Club School of Public Health and Primary Care, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China
- Microbiota I-Center (MagIC), Hong Kong, SAR, China
| | - Catherine J. Field
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Canada
| | - Matthew Hicks
- Edmonton Clinic Health Academy, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Jacqueline Pei
- Department of Educational Psychology, Faculty of Education, University of Alberta, Edmonton, Canada
| | - Anne Hicks
- Edmonton Clinic Health Academy, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Theo J. Moraes
- Hospital for Sick Children (SickKids), Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Elinor Simons
- Children's Hospital Research Institute of Manitoba, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada
| | - Stuart E. Turvey
- BC Children's Hospital, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Padmaja Subbarao
- Hospital for Sick Children (SickKids), Department of Pediatrics, University of Toronto, Toronto, Canada
- Dalla Lana School of Public Health, Division of Occupational and Environmental Health, University of Toronto, Toronto, Canada
| | - James A. Scott
- Dalla Lana School of Public Health, Division of Occupational and Environmental Health, University of Toronto, Toronto, Canada
| | - Piushkumar J. Mandhane
- Edmonton Clinic Health Academy, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Anita L. Kozyrskyj
- Edmonton Clinic Health Academy, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| |
Collapse
|
3
|
Charitos IA, Scacco S, Cotoia A, Castellaneta F, Castellana G, Pasqualotto F, Venneri M, Ferrulli A, Aliani M, Santacroce L, Carone M. Intestinal Microbiota Dysbiosis Role and Bacterial Translocation as a Factor for Septic Risk. Int J Mol Sci 2025; 26:2028. [PMID: 40076650 PMCID: PMC11900423 DOI: 10.3390/ijms26052028] [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: 11/19/2024] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
The human immune system is closely linked to microbiota such as a complex symbiotic relationship during the coevolution of vertebrates and microorganisms. The transfer of microorganisms from the mother's microbiota to the newborn begins before birth during gestation and is considered the initial phase of the intestinal microbiota (IM). The gut is an important site where microorganisms can establish colonies. The IM contains polymicrobial communities, which show complex interactions with diet and host immunity. The tendency towards dysbiosis of the intestinal microbiota is influenced by local but also extra-intestinal factors such as inflammatory processes, infections, or a septic state that can aggravate it. Pathogens could trigger an immune response, such as proinflammatory responses. In addition, changes in the host immune system also influence the intestinal community and structure with additional translocation of pathogenic and non-pathogenic bacteria. Finally, local intestinal inflammation has been found to be an important factor in the growth of pathogenic microorganisms, particularly in its role in sepsis. The aim of this article is to be able to detect the current knowledge of the mechanisms that can lead to dysbiosis of the intestinal microbiota and that can cause bacterial translocation with a risk of infection or septic state and vice versa.
Collapse
Affiliation(s)
- Ioannis Alexandros Charitos
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
- Doctoral School, Applied Neurosciences, University of Bari (UNIBA), 70124 Bari, Italy
| | - Salvatore Scacco
- Dipartimento di Biomedicina Traslazionale e Neuroscienze (DiBraiN), Scuola di Medicina, Università Degli Studi di Bari, Aldo Moro, 70124 Bari, Italy;
- U.O. Medicina, Ospedale Mater Dei-CBH, 70125 Bari, Italy
| | - Antonella Cotoia
- Department of Intensive Care, University Hospital of Foggia, 71121 Foggia, Italy
| | - Francesca Castellaneta
- U.O.C. Servizio di Immunoematologia e Medicina Trasfusionale—S.I.M.T. Ospedale Di Venere, 70131 Bari, Italy;
| | - Giorgio Castellana
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| | - Federico Pasqualotto
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
- Department of Public Health and Infectious Diseases, Pulmonary Division, Sapienza University of Rome, Policlinico Umberto I Hospital, Rome, Via del Policlinico 155, 00155 Rome, Italy
| | - Maria Venneri
- Genomics and Proteomics Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (M.V.); (A.F.)
| | - Angela Ferrulli
- Genomics and Proteomics Laboratory, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (M.V.); (A.F.)
| | - Maria Aliani
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, The University of Bari, 70124 Bari, Italy;
| | - Mauro Carone
- Pneumology and Respiratory Rehabilitation Unit, Istituti Clinici Scientifici Maugeri IRCCS, “Istitute” of Bari, 70124 Bari, Italy; (I.A.C.); (G.C.); (F.P.); (M.A.); (M.C.)
| |
Collapse
|
4
|
Artola-Borán M, Kirsche L, Fallegger A, Leary P, Tanriover M, Goodwin T, Geiger G, Hapfelmeier S, Yousefi S, Simon HU, Arnold IC, Müller A. IgA facilitates the persistence of the mucosal pathogen Helicobacter pylori. Mucosal Immunol 2025; 18:232-247. [PMID: 39581230 DOI: 10.1016/j.mucimm.2024.11.006] [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: 07/01/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/26/2024]
Abstract
IgA antibodies have an important role in clearing mucosal pathogens. In this study, we have examined the contribution of IgA to the immune control of the gastrointestinal bacterial pathogens Helicobacter pylori and Citrobacter rodentium. Both bacteria trigger a strong local IgA response that results in bacterial IgA coating in mice and in gastritis patients. Class switching to IgA depends on Peyer's patches, T-cells, eosinophils, and eosinophil-derived TGF-β in both models. In the case of H. pylori, IgA secretion and bacterial coating also depend on a functional bacterial type IV secretion system, which drives the generation of Th17 cells and the IL-17-dependent expression of the polymeric immunoglobulin receptor PIGR. IgA-/- mice are hypercolonized with C. rodentium in all examined tissues, suffer from more severe weight loss and develop more colitis. In contrast, H. pylori is controlled more efficiently in IgA-/- mice than their WT counterparts. The effects of IgA deficiency of the offspring can be compensated by maternal IgA delivered by WT foster mothers. We attribute the improved immune control observed in IgA-/- mice to IgA-mediated protection from complement killing, as H. pylori colonization is restored to wild type levels in a composite strain lacking both IgA and the central complement component C3. IgA antibodies can thus have protective or detrimental activities depending on the infectious agent.
Collapse
Affiliation(s)
- Mariela Artola-Borán
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Lydia Kirsche
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Angela Fallegger
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Peter Leary
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Functional Genomics Center Zürich, University of Zürich/ETHZ, Zürich, Switzerland
| | - Mine Tanriover
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Tanja Goodwin
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Gavin Geiger
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | | | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland; Institute of Biochemistry, Brandenburg Medical School, 16816 Neuruppin, Germany
| | - Isabelle C Arnold
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Comprehensive Cancer Center Zürich, Zürich, Switzerland.
| |
Collapse
|
5
|
Mohammadi A, Chiang S, Li F, Wei F, Lau CS, Aziz M, Ibarrondo FJ, Fulcher JA, Yang OO, Chia D, Kim Y, Wong DTW. Direct detection of 4-dimensions of SARS-CoV-2: infection (vRNA), infectivity (antigen), binding antibody, and functional neutralizing antibody in saliva. Sci Rep 2024; 14:30792. [PMID: 39730575 DOI: 10.1038/s41598-024-81019-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 11/22/2024] [Indexed: 12/29/2024] Open
Abstract
We developed a 4-parameter clinical assay using Electric Field Induced Release and Measurement (EFIRM) technology to simultaneously assess SARS-CoV-2 RNA (vRNA), nucleocapsid antigen, host binding (BAb) and neutralizing antibody (NAb) levels from a drop of saliva with performance that equals or surpasses current EUA-approved tests. The vRNA and antigen assays achieved lower limit of detection (LOD) of 100 copies/reaction and 3.5 TCID₅₀/mL, respectively. The vRNA assay differentiated between acutely infected (n = 10) and infection-naïve patients (n = 33) with an AUC of 0.9818, sensitivity of 90%, and specificity of 100%. The antigen assay similarly differentiated these patient populations with an AUC of 1.000. The BAb assay detected BAbs with an LOD of 39 pg/mL and distinguished acutely infected (n = 35), vaccinated with prior infection (n = 13), and vaccinated infection-naïve patients (n = 13) from pre-pandemic (n = 81) with AUC of 0.9481, 1.000, and 0.9962, respectively. The NAb assay detected NAbs with a LOD of 31.6 Unit/mL and differentiated between COVID-19 recovered or vaccinated patients (n = 31) and pre-pandemic controls (n = 60) with an AUC 0.923, sensitivity of 87.10%, and specificity of 86.67%. Our combo assay represents a significant technological advancement to simultaneously address SARS-CoV-2 infection and immunity, and it lays the foundation for tackling potential future pandemics.
Collapse
Grants
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
- U18 TR003778, U54 HL119893 NCATS NIH HHS
Collapse
Affiliation(s)
- Aida Mohammadi
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Samantha Chiang
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Feng Li
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Fang Wei
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | | | - Mohammad Aziz
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Francisco J Ibarrondo
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer A Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Otto O Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David Chia
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA
| | - Yong Kim
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA.
| | - David T W Wong
- School of Dentistry, University of California Los Angeles, 10833 Le Conte Ave., 73-022 CHS, Los Angeles, CA, 90095-1668, USA.
| |
Collapse
|
6
|
Jørgensen MR. Pathophysiological microenvironments in oral candidiasis. APMIS 2024; 132:956-973. [PMID: 38571459 DOI: 10.1111/apm.13412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/20/2024] [Indexed: 04/05/2024]
Abstract
Oral candidiasis (OC), a prevalent opportunistic infection of the oral mucosa, presents a considerable health challenge, particularly in individuals with compromised immune responses, advanced age, and local predisposing conditions. A considerable part of the population carries Candida in the oral cavity, but only few develop OC. Therefore, the pathogenesis of OC may depend on factors other than the attributes of the fungus, such as host factors and other predisposing factors. Mucosal trauma and inflammation compromise epithelial integrity, fostering a conducive environment for fungal invasion. Molecular insights into the immunocompromised state reveal dysregulation in innate and adaptive immunity, creating a permissive environment for Candida proliferation. Detailed examination of Candida species (spp.) and their virulence factors uncovers a nuanced understanding beyond traditional C. albicans focus, which embrace diverse Candida spp. and their strategies, influencing adhesion, invasion, immune evasion, and biofilm formation. Understanding the pathophysiological microenvironments in OC is crucial for the development of targeted therapeutic interventions. This review aims to unravel the diverse pathophysiological microenvironments influencing OC development focusing on microbial, host, and predisposing factors, and considers Candida resistance to antifungal therapy. The comprehensive approach offers a refined perspective on OC, seeking briefly to identify potential therapeutic targets for future effective management.
Collapse
Affiliation(s)
- Mette Rose Jørgensen
- Section of Oral Pathology and Oral Medicine, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Di Stefano M, Mirabella L, Cotoia A, Faleo G, Rauseo M, Rizzo AC, Fiore JR, Cinnella G, Serviddio G. A Possible Protective Effect of IgA Against Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) in Bronchoalveolar Lavage in COVID-19 Patients Admitted to Intensive Care Unit. Viruses 2024; 16:1851. [PMID: 39772161 PMCID: PMC11680283 DOI: 10.3390/v16121851] [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: 10/17/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
SARS-CoV-2 infection induces a humoral immune response, producing virus-specific antibodies such as IgM, IgG, and IgA. IgA antibodies are present at mucosal sites, protecting against respiratory and other mucosal infections, including SARS-CoV-2, by neutralizing viruses or impeding attachment to epithelial cells. Since SARS-CoV-2 spreads through the nasopharynx, the specific IgAs of SARS-CoV-2 are produced quickly after infection, effectively contributing to virus neutralization. Dimeric IgA has been reported to be 10 to 15 times more potent than its equivalent IgG, suggesting that this isotype may be particularly interesting in developing new monoclonal antibodies and/or new vaccines efficiently neutralizing the virus at the mucosal sites. It is still unclear whether IgA antibodies in BAL might play a role in the disease course and if their presence may have a prognostic significance. However, a harmful effect on diseases with high IgA titers has been reported. This study evaluated mucosal-specific IgA and IgG profiles in BAL of patients with COVID-19 acute respiratory failure admitted to the ICU. We included 57 patients (41 males and 16 females), admitted to the ICU of the University of Foggia. We used a commercially available ELISA assay to evaluate the presence of SARS-CoV-2 IgG and IgA antibodies in plasma and BAL of the 57 hospitalized patients with severe COVID-19 respiratory failure. However, 40/57 BAL and plasma from infected patients were available for the ELISA test; the remaining specimens were unsuitable. IgG and IgA antibodies against SARS-CoV-2 were detectable in 37 (92.5%) and 40 (100%) plasma specimens, respectively. IgG antibodies were found in a single sample, while IgAs were detected in 19 of 40 BAL samples analyzed. Correlations between these parameters and patient outcomes reveal a signature associated with survival. Interestingly, a statistically significant inverse correlation was found between the mortality rate and the presence of IgA to SARS-CoV-2 in BAL specimens. None of the 19 patients with a positive IgA died, compared to 7 out of 12 patients with a negative IgA-BAL (p: <0.0004). Despite being limited in size, this study suggests a significant protective effect of mucosal immunity in COVID-19 patients, even in advanced disease stages, and a role of IgA in the defense against the virus, as well as the possible use of effective vaccines and therapeutic strategies based on IgA antibodies.
Collapse
Affiliation(s)
- Mariantonietta Di Stefano
- Department of Surgical and Medical Science, Section of Infectious Diseases, University of Foggia, 71122 Foggia, Italy; (G.F.); (J.R.F.)
| | - Lucia Mirabella
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Antonella Cotoia
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Giuseppina Faleo
- Department of Surgical and Medical Science, Section of Infectious Diseases, University of Foggia, 71122 Foggia, Italy; (G.F.); (J.R.F.)
| | - Michela Rauseo
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Anna Chiara Rizzo
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Josè Ramon Fiore
- Department of Surgical and Medical Science, Section of Infectious Diseases, University of Foggia, 71122 Foggia, Italy; (G.F.); (J.R.F.)
| | - Gilda Cinnella
- Anesthesia and Intensive Care Unit, Department of Surgical and Medical Science, Policlinico Riuniti di Foggia, University of Foggia, 71122 Foggia, Italy; (L.M.); (A.C.); (M.R.); (A.C.R.); (G.C.)
| | - Gaetano Serviddio
- C.U.R.E. (University Center for Liver Disease Research and Treatment), Liver Unit, Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy;
| |
Collapse
|
8
|
Kim SJ, Lee HK, Kang KS, Lee MG, Shin MS. Korean Red Ginseng Polysaccharides Enhance Intestinal IgA Production and Barrier Function via Peyer's Patch Activation in Mice. Nutrients 2024; 16:3816. [PMID: 39599603 PMCID: PMC11597691 DOI: 10.3390/nu16223816] [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: 08/22/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Natural products are gaining attention for their potential benefits in gastrointestinal health. Plant-derived polysaccharides are essential for boosting intestinal immunity and maintaining gut homeostasis. This study investigated the effects of Korean red ginseng polysaccharides (KRG-P) on intestinal homeostasis including IgA and SCFA production and mucosal barrier integrity. Methods: Mice were orally administered KRG-P at doses of 50 mg/kg or 200 mg/kg for 10 days. Fecal IgA levels were measured on days 3, 5, and 11 and IgA from cultured Peyer's patch cells from KRG-P-treated mice were analyzed. Additionally, mRNA and protein expression levels of α-defensin, lysozyme, and E-cadherin in the small intestine were examined. Short-chain fatty acids (SCFAs) content in the cecum was also assessed. Results: KRG-P-treated groups showed a significant increase in fecal IgA levels on days 5 and 11, with no notable change on day 3. Cultured Peyer's patch cells from mice demonstrated heightened IgA production. Additionally, KRG-P administration upregulated α-defensin and lysozyme mRNA expression, along with elevated protein expression of E-cadherin, α-defensin, and lysozyme, in the small intestine. KRG-P treatment also led to increased cecal SCFA levels, including acetate, butyrate, and propionate. Conclusions: KRG-P may promote intestinal homeostasis and host defense mechanisms by activating immune cells in Peyer's patches, stimulating IgA production, enhancing antimicrobial peptide expression, and modulating gut microbiota metabolism through increased SCFA production.
Collapse
Affiliation(s)
- Sung Jin Kim
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.J.K.); (K.S.K.)
| | - Hae-Kyung Lee
- Avison Biomedical Research Center, Yonsei University, Seoul 03722, Republic of Korea;
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.J.K.); (K.S.K.)
| | - Mi-Gi Lee
- Bio-Center, Gyeonggi-do Business and Science Accelerator, Suwon 16229, Republic of Korea
| | - Myoung-Sook Shin
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea; (S.J.K.); (K.S.K.)
| |
Collapse
|
9
|
Yamasaki-Yashiki S, Shiraishi T, Gyobu M, Sasaki H, Kunisawa J, Yokota SI, Katakura Y. Immunostimulatory activity of lipoteichoic acid with three fatty acid residues derived from Limosilactobacillus antri JCM 15950 T. Appl Environ Microbiol 2024; 90:e0119724. [PMID: 39240119 PMCID: PMC11497808 DOI: 10.1128/aem.01197-24] [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: 06/27/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024] Open
Abstract
Some strains of lactic acid bacteria can regulate the host's intestinal immune system. Bacterial cells and membrane vesicles (MVs) of Limosilactobacillus antri JCM 15950T promote immunoglobulin A (IgA) production in murine Peyer's patch cells via toll-like receptor (TLR) 2. This study aimed to investigate the role of lipoteichoic acid (LTA), a ligand of TLR2, in the immunostimulatory activity of these bacterial cells and their MVs. LTA extracted from bacterial cells was purified through hydrophobic interaction chromatography and then divided into fractions LTA1 and LTA2 through anion-exchange chromatography. LTA1 induced greater interleukin (IL)-6 production from macrophage-like RAW264 cells than LTA2, and the induced IL-6 production was suppressed by TLR2 neutralization using an anti-TLR2 antibody. The LTAs in both fractions contained two hexose residues in the glycolipid anchor; however, LTA1 was particularly rich in triacyl LTA. The free hydroxy groups in the glycerol phosphate (GroP) repeating units were substituted by d-alanine (d-Ala) and α-glucose in LTA1, but only by α-glucose in LTA2. The dealanylation of LTA1 slightly suppressed IL-6 production in RAW264 cells, whereas deacylation almost completely suppressed IL-6 production. Furthermore, IL-6 production induced by dealanylated LTA1 was markedly higher than that induced by dealanylated LTA2. These results indicated that the critical moieties for the immunostimulatory activity of L. antri-derived LTA were the three fatty acid residues rather than the substitution with d-Ala in GroP. LTA was also detected in MVs, suggesting that the triacyl LTA, but not the diacyl LTA, translocated to the MVs and conferred immunostimulatory activity. IMPORTANCE Some lactic acid bacteria activate the host intestinal immune system via toll-like receptor (TLR) 2. Lipoteichoic acid (LTA) is a TLR2 ligand; however, the moieties of LTA that determine its immunostimulatory activity remain unclear because of the wide diversity of LTA partial structures. We found that Limosilactobacillus antri JCM 15950T has three types of LTAs (triacyl, diacyl, and monoacyl LTAs). Specifically, structural analysis of the LTAs revealed that triacyl LTA plays a crucial role in immunostimulation and that the fatty acid residues are essential for the activity. The three acyl residues are characteristic of LTAs from many lactic acid bacteria, and our findings can explain the immunostimulatory mechanisms widely exhibited by lactic acid bacteria. Furthermore, the immunostimulatory activity of membrane vesicles released by L. antri JCM 15950T is due to the transferred LTA, demonstrating a novel mechanism of membrane vesicle-mediated immunostimulation.
Collapse
Affiliation(s)
- Shino Yamasaki-Yashiki
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka, Japan
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health, and Nutrition, Ibaraki, Osaka, Japan
| | - Tsukasa Shiraishi
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Mai Gyobu
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka, Japan
| | - Haruna Sasaki
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials and Laboratory of Gut Environmental System, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health, and Nutrition, Ibaraki, Osaka, Japan
| | - Shin-ichi Yokota
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan
| | - Yoshio Katakura
- Department of Life Science and Biotechnology, Faculty of Chemistry, Materials and Bioengineering, Kansai University, Suita, Osaka, Japan
| |
Collapse
|
10
|
Gonzalez Agurto M, Olivares N, Canedo-Marroquin G, Espinoza D, Tortora SC. The Intersection of the Oral Microbiome and Salivary Metabolites in Head and Neck Cancer: From Diagnosis to Treatment. Cancers (Basel) 2024; 16:3545. [PMID: 39456639 PMCID: PMC11506592 DOI: 10.3390/cancers16203545] [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: 09/13/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Head and neck cancers (HNCs) are the seventh most common cancer worldwide, accounting for 4-5% of all malignancies. Salivary metabolites, which serve as key metabolic intermediates and cell-signalling molecules, are emerging as potential diagnostic biomarkers for HNC. While current research has largely concentrated on these metabolites as biomarkers, a critical gap remains in understanding their fluctuations before and after treatment, as well as their involvement in oral side effects. Recent studies emphasise the role of the oral microbiome and its metabolic activity in cancer progression and treatment efficacy by bacterial metabolites and virulence factors. Oral bacteria, such as P. gingivalis and F. nucleatum, contribute to a pro-inflammatory environment that promotes tumour growth. Additionally, F. nucleatum enhances its virulence through flagellar assembly and iron transport mechanisms, facilitating tumour invasion and survival. Moreover, alterations in the oral microbiome can influence chemotherapy efficacy and toxicity through the microbiota-host irinotecan axis, highlighting the complex interplay between microbial communities and therapeutic outcomes. Salivary metabolite profiles are influenced by factors such as gender, methods, and patient habits like smoking-a major risk factor for HNC. Radiotherapy (RT), a key treatment for HNC, often causes side effects such as xerostomia, oral mucositis, and swallowing difficulties which impact survivors' quality of life. Intensity-modulated radiotherapy (IMRT) aims to improve treatment outcomes and minimise side effects but can still lead to significant salivary gland dysfunction and associated complications. This review underscores the microbial and host interactions affecting salivary metabolites and their implications for cancer treatment and patient outcomes.
Collapse
Affiliation(s)
| | - Nicolas Olivares
- Faculty of Dentistry, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile;
| | - Gisela Canedo-Marroquin
- Faculty of Dentistry, Universidad de los Andes, Santiago 7620086, Chile;
- Faculty of Dentistry, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile;
- Millennium Institute on Immunology and Immunotherapy (MIII), Santiago 8331150, Chile
| | - Daniela Espinoza
- Faculty of Dentistry, Universidad Mayor, Santiago 8580745, Chile
| | - Sofia C. Tortora
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
11
|
Booth JS, Wahid R, Bruder D, Salerno-Goncalves R. Editorial: The synthesis of secretory immunoglobulin A in mucosal tissue: mucosal-associated invariant T, T follicular helper, and B cells. Front Immunol 2024; 15:1504432. [PMID: 39483477 PMCID: PMC11525977 DOI: 10.3389/fimmu.2024.1504432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024] Open
Affiliation(s)
- Jayaum S. Booth
- Center for Vaccine Development, University of Maryland, Baltimore, MD, United States
| | - Rezwanul Wahid
- Center for Vaccine Development, University of Maryland, Baltimore, MD, United States
| | - Dunja Bruder
- Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | | |
Collapse
|
12
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
- Ziyi Bai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
13
|
Bum Lee J, Huang Y, Oya Y, Nutzinger J, LE Ang Y, Sooi K, Chul Cho B, Soo RA. Modulating the gut microbiome in non-small cell lung cancer: Challenges and opportunities. Lung Cancer 2024; 194:107862. [PMID: 38959670 DOI: 10.1016/j.lungcan.2024.107862] [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] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/05/2024]
Abstract
Despite the efficacy of immunotherapy in non-small cell lung cancer (NSCLC), the majority of the patients experience relapse with limited subsequent treatment options. Preclinical studies of various epithelial tumors, such as melanoma and NSCLC, have shown that harnessing the gut microbiome resulted in improvement of therapeutic responses to immunotherapy. Is this review, we summarize the role of microbiome, including lung and gut microbiome in the context of NSCLC, provide overview of the mechanisms of microbiome in efficacy and toxicity of chemotherapies and immunotherapies, and address current ongoing clinical trials for NSCLC including fecal microbiota transplantation (FMT) and live biotherapeutic products (LBPs).
Collapse
Affiliation(s)
- Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yiqing Huang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Yuko Oya
- Department of Respiratory Medicine, Fujita Health University, Toyoake, Japan
| | - Jorn Nutzinger
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Yvonne LE Ang
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Kenneth Sooi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore.
| |
Collapse
|
14
|
Sam YH, Nibali L, Ghuman M. Periodontal granulation tissue - To remove or not to remove, that is the question. J Periodontal Res 2024; 59:636-646. [PMID: 38686698 DOI: 10.1111/jre.13261] [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: 08/01/2023] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 05/02/2024]
Abstract
Formation of granulation tissue is a fundamental phase in periodontal wound healing with subsequent maturation leading to regeneration or repair. However, persistently inflamed granulation tissue presents in osseous defects as a result of periodontitis and is routinely disrupted and discarded with non-surgical and surgical therapy to facilitate wound healing or improve chances of regeneration. Histological assessment suggests that granulation tissue from periodontitis-affected sites is effectively a chronic inflammatory tissue resulting from impaired wound healing due to persistence of bacterial dysbiotic bioflim. Nevertheless, the immunomodulatory potential and stem cell characteristics in granulation tissue have also raised speculation about the tissue's regenerative potential. This has led to the conception and recent implementation of surgical techniques which preserve granulation tissue with the intention of enhancing innate regenerative potential and improve clinical outcomes. As knowledge of fundamental cellular and molecular functions regulating periodontitis-affected granulation tissue is still scarce, this review aimed to provide a summary of current understanding of granulation tissue in the context of periodontal wound healing. This may provide new insights into clinical practice related to the management of granulation tissue and stimulate further investigation.
Collapse
Affiliation(s)
- Ye Han Sam
- Periodontology Unit, Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Luigi Nibali
- Periodontology Unit, Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Mandeep Ghuman
- Periodontology Unit, Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
15
|
Tobias J, Steinberger P, Wilkinson J, Klais G, Kundi M, Wiedermann U. SARS-CoV-2 Vaccines: The Advantage of Mucosal Vaccine Delivery and Local Immunity. Vaccines (Basel) 2024; 12:795. [PMID: 39066432 PMCID: PMC11281395 DOI: 10.3390/vaccines12070795] [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: 06/12/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Immunity against respiratory pathogens is often short-term, and, consequently, there is an unmet need for the effective prevention of such infections. One such infectious disease is coronavirus disease 19 (COVID-19), which is caused by the novel Beta coronavirus SARS-CoV-2 that emerged around the end of 2019. The World Health Organization declared the illness a pandemic on 11 March 2020, and since then it has killed or sickened millions of people globally. The development of COVID-19 systemic vaccines, which impressively led to a significant reduction in disease severity, hospitalization, and mortality, contained the pandemic's expansion. However, these vaccines have not been able to stop the virus from spreading because of the restricted development of mucosal immunity. As a result, breakthrough infections have frequently occurred, and new strains of the virus have been emerging. Furthermore, SARS-CoV-2 will likely continue to circulate and, like the influenza virus, co-exist with humans. The upper respiratory tract and nasal cavity are the primary sites of SARS-CoV-2 infection and, thus, a mucosal/nasal vaccination to induce a mucosal response and stop the virus' transmission is warranted. In this review, we present the status of the systemic vaccines, both the approved mucosal vaccines and those under evaluation in clinical trials. Furthermore, we present our approach of a B-cell peptide-based vaccination applied by a prime-boost schedule to elicit both systemic and mucosal immunity.
Collapse
Affiliation(s)
- Joshua Tobias
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Joy Wilkinson
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gloria Klais
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Kundi
- Department of Environmental Health, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
16
|
Pirker AL, Vogl T. Development of systemic and mucosal immune responses against gut microbiota in early life and implications for the onset of allergies. FRONTIERS IN ALLERGY 2024; 5:1439303. [PMID: 39086886 PMCID: PMC11288972 DOI: 10.3389/falgy.2024.1439303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024] Open
Abstract
The early microbial colonization of human mucosal surfaces is essential for the development of the host immune system. Already during pregnancy, the unborn child is prepared for the postnatal influx of commensals and pathogens via maternal antibodies, and after birth this protection is continued with antibodies in breast milk. During this critical window of time, which extends from pregnancy to the first year of life, each encounter with a microorganism can influence children's immune response and can have a lifelong impact on their life. For example, there are numerous links between the development of allergies and an altered gut microbiome. However, the exact mechanisms behind microbial influences, also extending to how viruses influence host-microbe interactions, are incompletely understood. In this review, we address the impact of infants' first microbial encounters, how the immune system develops to interact with gut microbiota, and summarize how an altered immune response could be implied in allergies.
Collapse
Affiliation(s)
| | - Thomas Vogl
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
17
|
Kumar Bharathkar S, Stadtmueller BM. Structural and Biochemical Requirements for Secretory Component Interactions with Dimeric IgA. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:226-234. [PMID: 38809110 PMCID: PMC11233122 DOI: 10.4049/jimmunol.2300717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
Secretory (S) IgA is the predominant mucosal Ab that protects host epithelial barriers and promotes microbial homeostasis. SIgA production occurs when plasma cells assemble two copies of monomeric IgA and one joining chain (JC) to form dimeric (d) IgA, which is bound by the polymeric Ig receptor (pIgR) on the basolateral surface of epithelial cells and transcytosed to the apical surface. There, pIgR is proteolytically cleaved, releasing SIgA, a complex of the dIgA and the pIgR ectodomain, called the secretory component (SC). The pIgR's five Ig-like domains (D1-D5) undergo a conformational change upon binding dIgA, ultimately contacting four IgA H chains and the JC in SIgA. In this study, we report structure-based mutational analysis combined with surface plasmon resonance binding assays that identify key residues in mouse SC D1 and D3 that mediate SC binding to dIgA. Residues in D1 CDR3 are likely to initiate binding, whereas residues that stabilize the D1-D3 interface are likely to promote the conformational change and stabilize the final SIgA structure. Additionally, we find that the JC's three C-terminal residues play a limited role in dIgA assembly but a significant role in pIgR/SC binding to dIgA. Together, these results inform models for the intricate mechanisms underlying IgA transport across epithelia and functions in the mucosa.
Collapse
Affiliation(s)
- Sonya Kumar Bharathkar
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
| | - Beth M. Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Carl R. Woese Institute of Genomic Biology
| |
Collapse
|
18
|
Withanage T, Lal M, Wachtel E, Patchornik G. Conjugated Nonionic Detergent Micelles: An Efficient Purification Platform for Dimeric Human Immunoglobulin A. ACS Med Chem Lett 2024; 15:979-986. [PMID: 38894919 PMCID: PMC11181477 DOI: 10.1021/acsmedchemlett.4c00128] [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: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/28/2024] [Indexed: 06/21/2024] Open
Abstract
The SARS-COV-2 virus is a deadly agent of inflammatory respiratory disease. Since 2020, studies have focused on developing new therapies based on galactose-rich IgA antibodies. Clinical surveys have also revealed that galactose-deficient IgA1 polymerizes in serum, producing IgA nephropathy, which is a common cause of kidney failure in young adults. Here we show that IgA1-IgA2 dimers are efficiently and economically purified in solution via conjugated nonionic surfactant micellar aggregates. Quantitative capture at pH 7 and extraction at pH 6.5 can avoid antibody exposure to acidic, potentially denaturing conditions. Brij-O20 aggregates lead to the highest process yields (88-91%) and purity (94%). Recovered IgA dimers preserve their native secondary structure and do not self-associate. Increasing the reaction volume has little impact on yield or purity. By introducing an efficient, inexpensive IgA purification protocol, we assist pharmaceutical firms and research laboratories in developing new IgA-based therapies as well as in increasing our understanding of IgA1 polymerization.
Collapse
Affiliation(s)
| | - Mitra Lal
- Department
of Chemical Sciences, Ariel University, 70400 Ariel, Israel
| | - Ellen Wachtel
- Faculty
of Chemistry, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Guy Patchornik
- Department
of Chemical Sciences, Ariel University, 70400 Ariel, Israel
| |
Collapse
|
19
|
Tang Y, Feng X, Lu Q, Cui C, Yu M, Wen Z, Luan Y, Dong L, Hu Z, Zhang R, Lu C, Liu J, Shinkura R, Hase K, Wang JY. MZB1-mediated IgA secretion suppresses the development and progression of colorectal cancer triggered by gut inflammation. Mucosal Immunol 2024; 17:450-460. [PMID: 38101774 DOI: 10.1016/j.mucimm.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/20/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Colorectal cancer (CRC) ranks among the top causes of mortality globally. Gut inflammation is one crucial risk factor that augments CRC development since patients suffering from inflammatory bowel disease have an increased incidence of CRC. The role of immunoglobulin (Ig)A in maintaining gut homeostasis and preventing inflammation has been well established. Our earlier work demonstrated that the marginal zone and B1 cell-specific protein (MZB1) promotes gut IgA secretion and its absence results in pronounced dextran sulfate sodium salt (DSS)-induced colitis. In the present study, we explored the role of MZB1 in CRC development using the azoxymethane (AOM)/DSS-induced CRC model. We observed an increase in both the number and size of the tumor nodules in Mzb1-/- mice compared with Mzb1+/+ mice. The increase in CRC development and progression in Mzb1-/- mice was associated with reduced intestinal IgA levels, altered gut flora, and more severe gut and systemic inflammation. Oral administration of the monoclonal IgA, W27, alleviated both the gut inflammation and AOM/DSS-induced CRC. Notably, cohousing Mzb1+/+ and Mzb1-/- mice from the 10th day after birth led to similar CRC development. Our findings underscore the pivotal role of MZB1-mediated IgA secretion in suppressing the onset and progression of CRC triggered by gut inflammation. Moreover, our study highlights the profound impact of microbiota composition, modulated by gut IgA levels, on gut inflammation. Nonetheless, establishing a direct correlation between the severity of colitis and subsequent CRC development and the presence or absence of a particular microbiota is challenging.
Collapse
Affiliation(s)
- Yue Tang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaoqian Feng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qing Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chaoqun Cui
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meiping Yu
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Zichao Wen
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yingying Luan
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lulu Dong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziying Hu
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Runyun Zhang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chunhui Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Reiko Shinkura
- Laboratory of Immunology and Infection Control, Institute of Quantitative Biosciences, the University of Tokyo, Tokyo, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China; Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Shanghai Sci-Tech Inno Center for Infection & Immunity, Shanghai 200052, China.
| |
Collapse
|
20
|
Kumar Bharathkar S, Stadtmueller BM. Structural and biochemical requirements for secretory component interactions with dimeric Immunoglobulin A. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.09.566401. [PMID: 38014291 PMCID: PMC10680632 DOI: 10.1101/2023.11.09.566401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Secretory (S) Immunoglobulin (Ig) A is the predominant mucosal antibody that protects host epithelial barriers and promotes microbial homeostasis. SIgA production occurs when plasma cells assemble two copies of monomeric IgA and one joining-chain (JC) to form dimeric (d) IgA, which is bound by the polymeric Ig receptor (pIgR) on the basolateral surface of epithelial cells and transcytosed to the apical surface. There, pIgR is proteolytically cleaved, releasing SIgA, a complex of the dIgA and the pIgR ectodomain, called secretory component (SC). The pIgR's five Ig-like domains (D1-D5) undergo a conformational change upon binding dIgA, ultimately contacting four IgA heavy chains and the JC in SIgA. Here we report structure-based mutational analysis combined with surface plasmon resonance binding assays that identify key residues in mouse SC D1 and D3 that mediate SC binding to dIgA. Residues in D1 CDR3 are likely to initiate binding whereas residues that stabilize the D1-D3 interface are likely to promote the conformation change and stabilize the final SIgA structure. Additionally, we find that the JC's three C-terminal residues play a limited role in dIgA assembly but a significant role in pIgR/SC binding to dIgA. Together results inform new models for the intricate mechanisms underlying IgA transport across epithelia and functions in the mucosa.
Collapse
Affiliation(s)
| | - Beth M. Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Carle R. Woese Institute of Genomic Biology
| |
Collapse
|
21
|
Bladh O, Aguilera K, Marking U, Kihlgren M, Greilert Norin N, Smed-Sörensen A, Sällberg Chen M, Klingström J, Blom K, Russell MW, Havervall S, Thålin C, Åberg M. Comparison of SARS-CoV-2 spike-specific IgA and IgG in nasal secretions, saliva and serum. Front Immunol 2024; 15:1346749. [PMID: 38558811 PMCID: PMC10978617 DOI: 10.3389/fimmu.2024.1346749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/30/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Several novel vaccine platforms aim at mucosal immunity in the respiratory tract to block SARS-CoV-2 transmission. Standardized methods for mucosal sample collection and quantification of mucosal antibodies are therefore urgently needed for harmonized comparisons and interpretations across mucosal vaccine trials and real-world data. Methods Using commercial electrochemiluminescence antibody panels, we compared SARS-CoV-2 spike-specific IgA and IgG in paired saliva, nasal secretions, and serum from 1048 healthcare workers with and without prior infection. Results Spike-specific IgA correlated well in nasal secretions and saliva (r>0.65, p<0.0001), but the levels were more than three-fold higher in nasal secretions as compared to in saliva (p<0.01). Correlations between the total population of spike-specific IgA and spike-specific secretory IgA (SIgA) were significantly stronger (p<0.0001) in nasal secretions (r=0.96, p<0.0001) as opposed to in saliva (r=0.77, p<0.0001), and spike-specific IgA correlated stronger (p<0.0001) between serum and saliva (r=0.73, p<0.001) as opposed to between serum and nasal secretions (r=0.54, p<0.001), suggesting transudation of monomeric spike specific IgA from the circulation to saliva. Notably, spike-specific SIgA had a markedly higher SARS-CoV-2 variant cross-binding capacity as compared to the total population of spike specific IgA and IgG in both nasal secretions, saliva and serum, (all p<0.0001), which emphasizes the importance of taking potential serum derived monomeric IgA into consideration when investigating mucosal immune responses. Discussion Taken together, although spike-specific IgA can be reliably measured in both nasal secretions and saliva, our findings imply an advantage of higher levels and likely also a larger proportion of SIgA in nasal secretions as compared to in saliva. We further corroborate the superior variant cross-binding capacity of SIgA in mucosal secretions, highlighting the potential protective benefits of a vaccine targeting the upper respiratory tract.
Collapse
Affiliation(s)
- Oscar Bladh
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Katherina Aguilera
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Marking
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Public Health Agency of Sweden, Solna, Sweden
| | - Martha Kihlgren
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Nina Greilert Norin
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Margaret Sällberg Chen
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Klingström
- Public Health Agency of Sweden, Solna, Sweden
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, Linköping, Sweden
| | - Kim Blom
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
- Public Health Agency of Sweden, Solna, Sweden
| | - Michael W. Russell
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Sebastian Havervall
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte Thålin
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry and SciLifeLab Affinity Proteomics, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Hu Y, Wu X, Zhou L, Liu J. Which is the optimal choice for neonates' formula or breast milk? NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:21. [PMID: 38488905 PMCID: PMC10942964 DOI: 10.1007/s13659-024-00444-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/28/2024] [Indexed: 03/17/2024]
Abstract
The incidence of prematurity has been increasing since the twenty-first century. Premature neonates are extremely vulnerable and require a rich supply of nutrients, including carbohydrates, proteins, docosahexaenoic acid (DHA), arachidonic acid (ARA) and others. Typical breast milk serves as the primary source for infants under six months old to provide these nutrients. However, depending on the individual needs of preterm infants, a more diverse and intricate range of nutrients may be necessary. This paper provides a comprehensive review of the current research progress on the physical and chemical properties, biological activity, function, and structure of breast milk, as well as explores the relationship between the main components of milk globular membrane and infant growth. Additionally, compare the nutritional composition of milk from different mammals and newborn milk powder, providing a comprehensive understanding of the differences in milk composition and detailed reference for meeting daily nutritional needs during lactation.
Collapse
Affiliation(s)
- Yueqi Hu
- National Demonstration Center for Experimental Ethnopharmacology Education, School of Pharmaceutical Sciences, South-Central MinZu University, Wuhan, 430074, People's Republic of China
| | - Xing Wu
- National Demonstration Center for Experimental Ethnopharmacology Education, School of Pharmaceutical Sciences, South-Central MinZu University, Wuhan, 430074, People's Republic of China
| | - Li Zhou
- National Demonstration Center for Experimental Ethnopharmacology Education, School of Pharmaceutical Sciences, South-Central MinZu University, Wuhan, 430074, People's Republic of China.
| | - Jikai Liu
- National Demonstration Center for Experimental Ethnopharmacology Education, School of Pharmaceutical Sciences, South-Central MinZu University, Wuhan, 430074, People's Republic of China.
| |
Collapse
|
23
|
Roja B, Chellapandi P. Design and characterization of a multi-epitope vaccine against Clostridium botulinum A3 Loch Maree intoxication in humans. Gene 2024; 892:147865. [PMID: 37783297 DOI: 10.1016/j.gene.2023.147865] [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: 06/23/2023] [Revised: 09/03/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023]
Abstract
Clostridium botulinum Loch Maree expresses an extremely potent botulinum neurotoxin subtype, A3 causing botulism and several gastrointestinal disorders in mammals. Several recombinant vaccines have been developed for human botulism and no vaccine is currently available for the treatment of diseases caused by other virulence factors. Hence, we designed, constructed, and characterized a multi-epitope vaccine from new virulence proteins identified from this organism using an immunoinformatics approach. The vaccine construct used in this study was designed from 6B cell linear epitopes, 12 cytotoxic T cell lymphocyte epitopes, and 15 helper T cell lymphocyte epitopes, with a defensin adjuvant and adjusting linker sequences. A molecular modeling approach was used to model, refine, and validate the 3D structure of the vaccine construct. Molecular docking studies were performed to determine the stability of the molecular interactions between the vaccine construct and human toll-like receptor 7. The in silico molecular cloning was used to clone a codon-optimized synthetic vaccine gene in pCYB1 vector and expressed in Escherichia coli. The results of this study identified six new virulence proteins: peptidoglycan hydrolase, SCP-like extracellular protein, N-acetylmuramoyl-l-alanine amidase, putative membrane protein, drug/metabolite exporter, and bacillolysin. The top B-cell, cytotoxic T-cell lymphocyte, and helper T-lymphocyte epitopes were predicted from these virulence proteins with greater accuracy and reliability. HLA-A*02:01 and HLA-A*03:01 were identified as HLA-A-binding alleles for cytotoxic T-cell lymphocyte epitopes. DRB1*0110 and DRB1*0115 are the dominant alleles that bind to helper T-cell lymphocyte epitopes. The synthetic gene construct was highly expressed in a heterologous host and produced considerable amounts of antigenic protein. The multi-epitope vaccine is more conservative in the sequence-structure-function link, immunogenic with less allergenicity, and possibly provokes cellular and humoral immunity. The present study suggests that the designed multi-epitope vaccine is a promising prophylactic candidate for the virulence and intoxication caused by subtype A3 strains.
Collapse
Affiliation(s)
- B Roja
- Industrial Systems Biology Lab, Department of Bioinformatics, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - P Chellapandi
- Industrial Systems Biology Lab, Department of Bioinformatics, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India.
| |
Collapse
|
24
|
Esmat K, Jamil B, Kheder RK, Kombe Kombe AJ, Zeng W, Ma H, Jin T. Immunoglobulin A response to SARS-CoV-2 infection and immunity. Heliyon 2024; 10:e24031. [PMID: 38230244 PMCID: PMC10789627 DOI: 10.1016/j.heliyon.2024.e24031] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 12/18/2023] [Accepted: 01/02/2024] [Indexed: 01/18/2024] Open
Abstract
The novel coronavirus disease (COVID-19) and its infamous "Variants" of the etiological agent termed Severe Acute Respiratory Syndrome Corona Virus 2 (SARS-CoV-2) has proven to be a global health concern. The three antibodies, IgA, IgM, and IgG, perform their dedicated role as main workhorses of the host adaptive immune system in virus neutralization. Immunoglobulin-A (IgA), also known as "Mucosal Immunoglobulin", has been under keen interest throughout the viral infection cycle. Its importance lies because IgA is predominant mucosal antibody and SARS family viruses primarily infect the mucosal surfaces of human respiratory tract. Therefore, IgA can be considered a diagnostic and prognostic marker and an active infection biomarker for SARS CoV-2 infection. Along with molecular analyses, serological tests, including IgA detection tests, are gaining ground in application as an early detectable marker and as a minimally invasive detection strategy. In the current review, it was emphasized the role of IgA response in diagnosis, host defense strategies, treatment, and prevention of SARS-CoV-2 infection. The data analysis was performed through almost 100 published peer-reviewed research reports and comprehended the importance of IgA in antiviral immunity against SARS-CoV-2 and other related respiratory viruses. Taken together, it is concluded that secretory IgA- Abs can serve as a promising detection tool for respiratory viral diagnosis and treatment parallel to IgG-based therapeutics and diagnostics. Vaccine candidates that target and trigger mucosal immune response may also be employed in future dimensions of research against other respiratory viruses.
Collapse
Affiliation(s)
- Khaleqsefat Esmat
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Baban Jamil
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, KRG, Erbil, Iraq
| | - Ramiar Kaml Kheder
- Medical Laboratory Science Department, College of Science, University of Raparin, Rania, Sulaymaniyah, Iraq
| | - Arnaud John Kombe Kombe
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Weihong Zeng
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Huan Ma
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
| | - Tengchuan Jin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Laboratory of Structural Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, Anhui, 230027, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
| |
Collapse
|
25
|
Dinis M, Tran NC. Oral immune system and microbes. MICROBES, MICROBIAL METABOLISM, AND MUCOSAL IMMUNITY 2024:147-228. [DOI: 10.1016/b978-0-323-90144-4.00005-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Zurutuza JI, Gonzalez S, Calderón AL, Caba M, Ramos FR. Changes in the Immunology of Breast Milk From Obese or Overweight Women: a Brief Review. Cureus 2024; 16:e52207. [PMID: 38327967 PMCID: PMC10847007 DOI: 10.7759/cureus.52207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2024] [Indexed: 02/09/2024] Open
Abstract
A systematic search was carried out through search platforms and specialized databases, such as Academic Google, PubMed, and Scopus, using thesauri: breast feeding, obesity, immunology, and human milk in English and Spanish, and those articles published from January 2000 to December 2021, in both languages. Only those reports that included quantitative data on immunological components in the milk of normal-weight and overweight women were considered. The PRISMA 2020 guides were used, and a total of 306 articles were reviewed, of which a total of 33 were included, according to the basic inclusion criteria. It was observed that in obese mothers, there is an increase in certain immune cells, such as B lymphocytes, cytotoxic T lymphocytes, and NK cells, and cytokines, such as IL-6 and IFN-γ; other alterations included the bacterial population and proteins with antibacterial action. Also, a decrease in growth factors such as TGF-β and IFG-1 was documented in overweight women. Immunoglobulin concentrations did not show substantial changes. This brief review shows that maternal overweight is associated with changes in the biochemical and immunological parameters of milk.
Collapse
Affiliation(s)
- Jorge I Zurutuza
- Epidemiology and Biostatistics, Centro de Investigaciones Biomedicas, Universidad Veracruzana, Xalapa, MEX
| | - Santiago Gonzalez
- Chemistry, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, MEX
| | - Ana L Calderón
- Neuropathology, Instituto Nacional de Neurología y Neurocirugia, Ciudad de Mexico, MEX
| | - Mario Caba
- Neuroscience, Centro de Investigaciones Biomédicas, Universidad Veracruzana, Xalapa, MEX
| | - Fernando R Ramos
- Chemistry, Instituto de Quimica Aplicada, Universidad Veracruzana, Xalapa, MEX
| |
Collapse
|
27
|
Shinkura R. Development of Orally Ingestible IgA Antibody Drugs to Maintain Symbiosis Between Humans and Microorganisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:165-176. [PMID: 38467979 DOI: 10.1007/978-981-99-9781-7_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
In recent years, dysbiosis, abnormalities in the gut microbiota, has been reported to be associated with the development of many diseases, and improving the gut microbiota is important for health maintenance. It has been shown that the host recognizes and regulates intestinal bacteria by means of IgA antibodies secreted into the gut, but the precise nature of the commensal gut bacteria recognized by each IgA antibody is unclear. We have cloned monoclonal IgA antibodies from mouse intestinal IgA-producing cells and are searching for bacterial molecules recognized by each IgA clone. Although the interaction of IgA antibodies with intestinal bacteria is still largely unknown and requires further basic research, we discuss the potential use of orally ingestible IgA antibodies as agents to improve intestinal microbiota.
Collapse
Affiliation(s)
- Reiko Shinkura
- Laboratory of Immunology and Infection Control, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
28
|
Xu X, Delves PJ, Huang J, Shao W, Qiu X. Comparison of Non B-Ig and B-Ig. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1445:73-88. [PMID: 38967751 DOI: 10.1007/978-981-97-0511-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
Immunoglobulin (Ig) has been widely acknowledged to be produced solely by B-lineage cells. However, growing evidence has demonstrated the expression of Ig in an array of cancer cells, as well as normal cells including epithelial cells, epidermal cells, mesangial cells, monocytes, and neutrophils. Ig has even been found to be expressed in non-B cells at immune-privileged sites such as neurons and spermatogenic cells. Despite these non-B cell-derived Igs (non-B-Igs) sharing the same symmetric structures with conventional Igs (B-Igs), further studies have revealed unique characteristics of non-B-Ig, such as restricted variable region and aberrant glycosylation. Moreover, non-B-Ig exhibits properties of promoting malignant behaviours of cancer cells, therefore it could be utilised in the clinic as a potential therapeutic biomarker or target. The elucidation of the generation and regulation of non-B-Ig will certainly broaden our understanding of immunology.
Collapse
Affiliation(s)
- Xiaojun Xu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Peter J Delves
- Division of Infection and Immunity, Department of Immunology, UCL (University College London), London, UK
| | - Jing Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Xiaoyan Qiu
- Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
29
|
Mohammadi A, Chiang S, Li F, Wei F, Lau CS, Aziz M, Ibarrondo FJ, Fulcher JA, Yang OO, Chia D, Kim Y, Wong DT. Direct Detection of 4-Dimensions of SARS-CoV-2: Infection (vRNA), Infectivity (Antigen), Binding Antibody, and Functional Neutralizing Antibody in Saliva. RESEARCH SQUARE 2023:rs.3.rs-3745787. [PMID: 38234820 PMCID: PMC10793499 DOI: 10.21203/rs.3.rs-3745787/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
We developed a 4-parameter clinical assay using Electric Field Induced Release and Measurement (EFIRM) technology to simultaneously assess SARS-CoV-2 RNA (vRNA), nucleocapsid antigen, host binding (BAb) and neutralizing antibody (NAb) levels from a drop of saliva with performance that equals or surpasses current EUA-approved tests. The vRNA and antigen assays achieved lower limit of detection (LOD) of 100 copies/reaction and 3.5 TCID₅₀/mL, respectively. The vRNA assay differentiated between acutely infected (n=10) and infection-naïve patients (n=33) with an AUC of 0.9818, sensitivity of 90%, and specificity of 100%. The antigen assay similarly differentiated these patient populations with an AUC of 1.000. The BAb assay detected BAbs with an LOD of 39 pg/mL and distinguished acutely infected (n=35), vaccinated with prior infection (n=13), and vaccinated infection-naïve patients (n=13) from control (n=81) with AUC of 0.9481, 1.000, and 0.9962, respectively. The NAb assay detected NAbs with an LOD of 31.6 Unit/mL and differentiated between COVID-19 recovered or vaccinated patients (n=31) and pre-pandemic controls (n=60) with an AUC 0.923, sensitivity of 87.10%, and specificity of 86.67%. Our multiparameter assay represents a significant technological advancement to simultaneously address SARS-CoV-2 infection and immunity, and it lays the foundation for tackling potential future pandemics.
Collapse
Affiliation(s)
- Aida Mohammadi
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Samantha Chiang
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Feng Li
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Fang Wei
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Mohammad Aziz
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Francisco J. Ibarrondo
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer A. Fulcher
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Otto O. Yang
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David Chia
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - Yong Kim
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| | - David T.W. Wong
- School of Dentistry, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
30
|
Gambirasi M, Safa A, Vruzhaj I, Giacomin A, Sartor F, Toffoli G. Oral Administration of Cancer Vaccines: Challenges and Future Perspectives. Vaccines (Basel) 2023; 12:26. [PMID: 38250839 PMCID: PMC10821404 DOI: 10.3390/vaccines12010026] [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: 11/23/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Cancer vaccines, a burgeoning strategy in cancer treatment, are exploring innovative administration routes to enhance patient and medical staff experiences, as well as immunological outcomes. Among these, oral administration has surfaced as a particularly noteworthy approach, which is attributed to its capacity to ignite both humoral and cellular immune responses at systemic and mucosal tiers, thereby potentially bolstering vaccine efficacy comprehensively and durably. Notwithstanding this, the deployment of vaccines through the oral route in a clinical context is impeded by multifaceted challenges, predominantly stemming from the intricacy of orchestrating effective oral immunogenicity and necessitating strategic navigation through gastrointestinal barriers. Based on the immunogenicity of the gastrointestinal tract, this review critically analyses the challenges and recent advances and provides insights into the future development of oral cancer vaccines.
Collapse
Affiliation(s)
- Marta Gambirasi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Amin Safa
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Doctoral School in Pharmacological Sciences, University of Padua, 35131 Padova, Italy
- Department of Immunology, School of Medicine, Zabol University of Medical Sciences, Zabol 98616-15881, Iran
| | - Idris Vruzhaj
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
- Doctoral School in Pharmacological Sciences, University of Padua, 35131 Padova, Italy
| | - Aurora Giacomin
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Franca Sartor
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS National Cancer Institute, 33081 Aviano, Italy; (M.G.); (I.V.); (F.S.)
| |
Collapse
|
31
|
Bertin B, Foligne B, Ley D, Lesage J, Beghin L, Morcel J, Gottrand F, Hermann E. An Overview of the Influence of Breastfeeding on the Development of Inflammatory Bowel Disease. Nutrients 2023; 15:5103. [PMID: 38140362 PMCID: PMC10745409 DOI: 10.3390/nu15245103] [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: 11/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The first 1000 days of life is a critical period that contributes significantly to the programming of an individual's future health. Among the many changes that occur during this period early in life, there is growing evidence that the establishment of healthy gut microbiota plays an important role in the prevention of both short- and long-term health problems. Numerous publications suggest that the quality of the gut microbiota colonisation depends on several dietary factors, including breastfeeding. In this respect, a relationship between breastfeeding and the risk of inflammatory bowel disease (IBD) has been suggested. IBDs are chronic intestinal diseases, and perinatal factors may be partly responsible for their onset. We review the existence of links between breastfeeding and IBD based on experimental and clinical studies. Overall, despite encouraging experimental data in rodents, the association between breastfeeding and the development of IBD remains controversial in humans, partly due to the considerable heterogeneity between clinical studies. The duration of exclusive breastfeeding is probably decisive for its lasting effect on IBD. Thus, specific improvements in our knowledge could support dietary interventions targeting the gut microbiome, such as the early use of prebiotics, probiotics or postbiotics, in order to prevent the disease.
Collapse
Affiliation(s)
- Benjamin Bertin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Benoit Foligne
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Laurent Beghin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Jules Morcel
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| |
Collapse
|
32
|
Conca W, Saleh SM, Al-Rabiah R, Parhar RS, Abd-Elnaeim M, Al-Hindas H, Tinson A, Kroell KB, Liedl KR, Collison K, Kishore U, Al-Mohanna F. The immunoglobulin A isotype of the Arabian camel ( Camelus dromedarius) preserves the dualistic structure of unconventional single-domain and canonical heavy chains. Front Immunol 2023; 14:1289769. [PMID: 38162642 PMCID: PMC10756906 DOI: 10.3389/fimmu.2023.1289769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/30/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction The evolution of adaptive immunity in Camelidae resulted in the concurrent expression of classic heterotetrameric and unconventional homodimeric heavy chain-only IgG antibodies. Heavy chain-only IgG bears a single variable domain and lacks the constant heavy (CH) γ1 domain required for pairing with the light chain. It has not been reported whether this distinctive feature of IgG is also observed in the IgA isotype. Methods Gene-specific primers were used to generate an IgA heavy chain cDNA library derived from RNA extracted from the dromedary's third eyelid where isolated lymphoid follicles and plasma cells abound at inductive and effector sites, respectively. Results Majority of the cDNA clones revealed hallmarks of heavy chain-only antibodies, i.e. camelid-specific amino acid substitutions in framework region 1 and 2, broad length distribution of complementarity determining region 3, and the absence of the CHα1 domain. In a few clones, however, the cDNA of the canonical IgA heavy chain was amplified which included the CHα1 domain, analogous to CHγ1 domain in IgG1 subclass. Moreover, we noticed a short, proline-rich hinge, and, at the N-terminal end of the CHα3 domain, a unique, camelid-specific pentapeptide of undetermined function, designated as the inter-α region. Immunoblots using rabbit anti-camel IgA antibodies raised against CHα2 and CHα3 domains as well as the inter-α region revealed the expression of a ~52 kDa and a ~60 kDa IgA species, corresponding to unconventional and canonical IgA heavy chain, respectively, in the third eyelid, trachea, small and large intestine. In contrast, the leporine anti-CHα1 antibody detected canonical, but not unconventional IgA heavy chain, in all the examined tissues, milk, and serum, in addition to another hitherto unexplored species of ~45 kDa in milk and serum. Immunohistology using anti-CHα domain antibodies confirmed the expression of both variants of IgA heavy chains in plasma cells in the third eyelid's lacrimal gland, conjunctiva, tracheal and intestinal mucosa. Conclusion We found that in the dromedary, the IgA isotype has expanded the immunoglobulin repertoire by co-expressing unconventional and canonical IgA heavy chains, comparable to the IgG class, thus underscoring the crucial role of heavy chain-only antibodies not only in circulation but also at the mucosal frontiers.
Collapse
Affiliation(s)
- Walter Conca
- Department of Executive Health Medicine, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Soad M. Saleh
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Rana Al-Rabiah
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Ranjit Singh Parhar
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Mahmoud Abd-Elnaeim
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Hussein Al-Hindas
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Alexander Tinson
- Management of Scientific Centers and Presidential Camels, Department of President’s Affairs, Hilli ET and Cloning Centre, Al Ain, United Arab Emirates
| | | | - Klaus Roman Liedl
- Center for Chemistry and Biomedicine, University of Innsbruck, Innsbruck, Austria
| | - Kate Collison
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Uday Kishore
- Department of Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Futwan Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
33
|
Kuthyar S, Diaz J, Avalos-Villatoro F, Maltecca C, Tiezzi F, Dunn RR, Reese AT. Domestication shapes the pig gut microbiome and immune traits from the scale of lineage to population. J Evol Biol 2023; 36:1695-1711. [PMID: 37885134 DOI: 10.1111/jeb.14227] [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: 01/24/2023] [Revised: 08/22/2023] [Accepted: 08/29/2023] [Indexed: 10/28/2023]
Abstract
Animal ecology and evolution have long been known to shape host physiology, but more recently, the gut microbiome has been identified as a mediator between animal ecology and evolution and health. The gut microbiome has been shown to differ between wild and domestic animals, but the role of these differences for domestic animal evolution remains unknown. Gut microbiome responses to new animal genotypes and local environmental change during domestication may promote specific host phenotypes that are adaptive (or not) to the domestic environment. Because the gut microbiome supports host immune function, understanding the effects of animal ecology and evolution on the gut microbiome and immune phenotypes is critical. We investigated how domestication affects the gut microbiome and host immune state in multiple pig populations across five domestication contexts representing domestication status and current living conditions: free-ranging wild, captive wild, free-ranging domestic, captive domestic in research or industrial settings. We observed that domestication context explained much of the variation in gut microbiome composition, pathogen abundances and immune markers, yet the main differences in the repertoire of metabolic genes found in the gut microbiome were between the wild and domestic genetic lineages. We also documented population-level effects within domestication contexts, demonstrating that fine scale environmental variation also shaped host and microbe features. Our findings highlight that understanding which gut microbiome and immune traits respond to host genetic lineage and/or scales of local ecology could inform targeted interventions that manipulate the gut microbiome to achieve beneficial health outcomes.
Collapse
Affiliation(s)
- Sahana Kuthyar
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
| | - Jessica Diaz
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
| | | | - Christian Maltecca
- Department of Animal Science, North Carolina State University, Raleigh, North Carolina, USA
| | - Francesco Tiezzi
- Department of Agriculture, Food, Environment and Forestry, University of Florence, Florence, Italy
| | - Robert R Dunn
- Department of Applied Ecology, North Carolina State University, Raleigh, North Carolina, USA
| | - Aspen T Reese
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
34
|
Bharathkar SK, Miller MJ, Stadtmueller BM. Engineered Secretory Immunoglobulin A provides insights on antibody-based effector mechanisms targeting Clostridiodes difficile. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566291. [PMID: 37986930 PMCID: PMC10659285 DOI: 10.1101/2023.11.08.566291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Secretory (S) Immunoglobin (Ig) A is the predominant mucosal antibody, which mediates host interactions with commensal and pathogenic microbes, including Clostridioides difficile. SIgA adopts a polymeric IgA structure that is bound by secretory component (SC). Despite significance, how SIgA supports diverse effector mechanisms is poorly characterized and SIgA-based therapies nonexistent. We engineered chimeric (c) SIgAs, in which we replaced SC domain D2 with a single domain antibody or a monomeric fluorescent protein, allowing us to investigate and enhance SIgA effector mechanisms. cSIgAs exhibited increased neutralization potency against C. difficile toxins, promoted bacterial clumping and cell rupture, and decreased cytotoxicity. cSIgA also allowed us to visualize and/or quantify C. difficile morphological changes and clumping events. Results reveal mechanisms by which SIgA combats C. difficile infection, demonstrate that cSIgA design can modulate these mechanisms, and demonstrate cSIgA's adaptability to modifications that might target a broad range of antigens and effector mechanisms.
Collapse
Affiliation(s)
- Sonya Kumar Bharathkar
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
| | - Michael J. Miller
- Carle R. Woese Institute of Genomic Biology
- Department of food science and Human Nutrition, University of Illinois Urbana-Champaign, Illinois 61801 USA
| | - Beth M. Stadtmueller
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Department of Biomedical and Translational Sciences, Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, Illinois 61801 USA
- Carle R. Woese Institute of Genomic Biology
| |
Collapse
|
35
|
Li JP, Du YT, Guo C, Rao XR, Li S. IgA nephropathy to proliferative glomerulonephritis with monoclonal IgAκ deposits: a pattern switch. J Nephrol 2023; 36:2375-2380. [PMID: 36913081 PMCID: PMC10638190 DOI: 10.1007/s40620-023-01583-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 01/13/2023] [Indexed: 03/14/2023]
Abstract
We report the case of a 31-year-old male who presented with repeated episodes of nephritic-nephrotic syndrome in concomitance with infection. IgA was diagnosed and was initially responsive to treatment with immunosuppressors but further disease flare did not respond to treatment. Based on three consecutive renal biopsies over 8 years, a pattern switch from endocapillary proliferative IgA nephropathy to membranous proliferative glomerulonephritis with monoclonal IgAκ deposits was observed. Bortezomib-dexamethasone combination therapy finally led to a favorable renal response. This case provides new insights into the pathophysiological mechanisms of proliferative glomerulonephritis with monoclonal immunoglobin deposits (PGNMID), highlighting the importance of repeat renal biopsies and routine evaluation of monoclonal immunoglobin deposits in proliferative glomerulonephritis with refractory nephrotic syndrome.
Collapse
Affiliation(s)
- Jin-Pu Li
- Renal Division, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange St. Xicheng District, Beijing, People's Republic of China
| | - Ya-Ting Du
- Renal Division, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange St. Xicheng District, Beijing, People's Republic of China
| | - Chuan Guo
- Renal Division, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange St. Xicheng District, Beijing, People's Republic of China
| | - Xiang-Rong Rao
- Renal Division, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange St. Xicheng District, Beijing, People's Republic of China.
| | - Shen Li
- Renal Division, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5 Beixiange St. Xicheng District, Beijing, People's Republic of China.
| |
Collapse
|
36
|
van Dalen R, Elsherbini AMA, Harms M, Alber S, Stemmler R, Peschel A. Secretory IgA impacts the microbiota density in the human nose. MICROBIOME 2023; 11:233. [PMID: 37865781 PMCID: PMC10589987 DOI: 10.1186/s40168-023-01675-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 09/24/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND Respiratory mucosal host defense relies on the production of secretory IgA (sIgA) antibodies, but we currently lack a fundamental understanding of how sIgA is induced by contact with microbes and how such immune responses may vary between humans. Defense of the nasal mucosal barrier through sIgA is critical to protect from infection and to maintain homeostasis of the microbiome, which influences respiratory disorders and hosts opportunistic pathogens. METHODS We applied IgA-seq analysis to nasal microbiota samples from male and female healthy volunteers, to identify which bacterial genera and species are targeted by sIgA on the level of the individual host. Furthermore, we used nasal sIgA from the same individuals in sIgA deposition experiments to validate the IgA-seq outcomes. CONCLUSIONS We observed that the amount of sIgA secreted into the nasal mucosa by the host varied substantially and was negatively correlated with the bacterial density, suggesting that nasal sIgA limits the overall bacterial capacity to colonize. The interaction between mucosal sIgA antibodies and the nasal microbiota was highly individual with no obvious differences between potentially invasive and non-invasive bacterial species. Importantly, we could show that for the clinically relevant opportunistic pathogen and frequent nasal resident Staphylococcus aureus, sIgA reactivity was in part the result of epitope-independent interaction of sIgA with the antibody-binding protein SpA through binding of sIgA Fab regions. This study thereby offers a first comprehensive insight into the targeting of the nasal microbiota by sIgA antibodies. It thereby helps to better understand the shaping and homeostasis of the nasal microbiome by the host and may guide the development of effective mucosal vaccines against bacterial pathogens. Video Abstract.
Collapse
Affiliation(s)
- Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany.
- Present Address: Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - Ahmed M A Elsherbini
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Mareike Harms
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Svenja Alber
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Regine Stemmler
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, Department of Infection Biology, University of Tübingen, Tübingen, Germany.
- Cluster of Excellence EXC2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany.
- German Center for Infection Research (DZIF), Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
37
|
Mahdally SM, Izquierdo M, Viscardi RM, Magder LS, Crowley HM, Bafford AC, Drachenberg CB, Farfan MJ, Fasano A, Sztein MB, Salerno-Goncalves R. Secretory-IgA binding to intestinal microbiota attenuates inflammatory reactions as the intestinal barrier of preterm infants matures. Clin Exp Immunol 2023; 213:339-356. [PMID: 37070830 PMCID: PMC10570995 DOI: 10.1093/cei/uxad042] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/09/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023] Open
Abstract
Previous work has shown that Secretory-IgA (SIgA) binding to the intestinal microbiota is variable and may regulate host inflammatory bowel responses. Nevertheless, the impact of the SIgA functional binding to the microbiota remains largely unknown in preterm infants whose immature epithelial barriers make them particularly susceptible to inflammation. Here, we investigated SIgA binding to intestinal microbiota isolated from stools of preterm infants <33 weeks gestation with various levels of intestinal permeability. We found that SIgA binding to intestinal microbiota attenuates inflammatory reactions in preterm infants. We also observed a significant correlation between SIgA affinity to the microbiota and the infant's intestinal barrier maturation. Still, SIgA affinity was not associated with developing host defenses, such as the production of mucus and inflammatory calprotectin protein, but it depended on the microbiota shifts as the intestinal barrier matures. In conclusion, we reported an association between the SIgA functional binding to the microbiota and the maturity of the preterm infant's intestinal barrier, indicating that the pattern of SIgA coating is altered as the intestinal barrier matures.
Collapse
Affiliation(s)
- Sarah M Mahdally
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mariana Izquierdo
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rose M Viscardi
- Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Laurence S Magder
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Helena M Crowley
- Division of Pediatric Surgery and Urology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andrea C Bafford
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cinthia B Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mauricio J Farfan
- Departamento de Pediatría y Cirugía Infantil, Facultad de Medicina, Hospital Dr. Luis Calvo Mackenna, Universidad de Chile, Santiago, Chile
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Rosangela Salerno-Goncalves
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
38
|
Gaglio SC, Perduca M, Zipeto D, Bardi G. Efficiency of Chitosan Nanocarriers in Vaccinology for Mucosal Immunization. Vaccines (Basel) 2023; 11:1333. [PMID: 37631901 PMCID: PMC10459455 DOI: 10.3390/vaccines11081333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/29/2023] Open
Abstract
The mucosal barrier constitutes a huge surface area, close to 40 m2 in humans, located mostly in the respiratory, gastrointestinal and urogenital tracts and ocular cavities. It plays a crucial role in tissue interactions with the microbiome, dietary antigens and other environmental materials. Effective vaccinations to achieve highly protective mucosal immunity are evolving strategies to counteract several serious diseases including tuberculosis, diphtheria, influenzae B, severe acute respiratory syndrome, Human Papilloma Virus infection and Acquired Immune Deficiency Syndrome. Interestingly, one of the reasons behind the rapid spread of severe acute respiratory syndrome coronavirus 2 variants has been the weakness of local immunization at the level of the respiratory mucosa. Mucosal vaccines can outperform parenteral vaccination as they specifically elicit protective mucosal immune responses blocking infection and transmission. In this scenario, chitosan-based nanovaccines are promising adjuvants-carrier systems that rely on the ability of chitosan to cross tight junctions and enhance particle uptake due to chitosan-specific mucoadhesive properties. Indeed, chitosan not only improves the adhesion of antigens to the mucosa promoting their absorption but also shows intrinsic immunostimulant abilities. Furthermore, by finely tuning the colloidal properties of chitosan, it can provide sustained antigen release to strongly activate the humoral defense. In the present review, we agnostically discuss the potential reasons why chitosan-based vaccine carriers, that efficiently elicit strong immune responses in experimental setups and in some pre-clinical/clinical studies, are still poorly considered for therapeutic formulations.
Collapse
Affiliation(s)
- Salvatore Calogero Gaglio
- Department of Biotechnology, University of Verona, Cà Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy;
| | - Massimiliano Perduca
- Department of Biotechnology, University of Verona, Cà Vignal 1, Strada Le Grazie 15, 37134 Verona, Italy;
| | - Donato Zipeto
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy
| | - Giuseppe Bardi
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
39
|
Castilla Gómez de Agüero V, Valderas-García E, González Del Palacio L, Giráldez FJ, Balaña-Fouce R, Martínez-Valladares M. Secretory IgA as Biomarker for Gastrointestinal Nematodes Natural Infection in Different Breed Sheep. Animals (Basel) 2023; 13:2189. [PMID: 37443987 DOI: 10.3390/ani13132189] [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: 05/30/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Specific IgA antibody has been shown to play an important role in resistance to gastrointestinal nematode (GIN) infections in sheep, particularly in Teladorsagia circumcincta parasitosis. In some breeds, negative associations have been shown between IgA levels and worm burden in experimentally infected sheep. In the present study, we have studied the relationship between IgA levels in naturally infected sheep (582 ewes in total; 193 younger than one year old and 389 older than one year old) and fecal egg count (FEC) in the Assaf, Castellana, and Churra breeds. ELISA assays were performed to measure IgA levels against the somatic antigen of T. circumcincta third larval stage (L3) and a 203-amino-acid fragment of the protein disulfide isomerase from the same GIN species. A multilevel random intercept model was developed to predict the infection risk according to age or breed. Spearman's correlation rank was used for statistical analysis. The prediction model showed that breed was not an influential factor in this study, although the Assaf breed could be considered slightly more susceptible than the others. In addition, age affected the infection risk, with the young ewes more susceptible to infection than the adult groups, except for the Castellana breed, whose risk of infection was similar at all ages. The most significant positive association was found between FEC and IgA measured in the nasal secretions of young ewes using both antigens (Rho = 0.5; p = 0.00); the correlation of FEC with IgA in serum was moderately significant (Rho = 0.306; p = 0.00). Comparing both antigens, the protein disulfide isomerase antigen was less reactive than the somatic antigen from L3. In conclusion, under natural conditions, specific IgA against GIN was positively associated with FEC in sheep, with nasal secretions from young animals being the sample where this association is stronger, which, therefore, could be used as a marker of infection in further studies.
Collapse
Affiliation(s)
- Verónica Castilla Gómez de Agüero
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, 24346 Grulleros, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, 24007 León, Spain
| | - Elora Valderas-García
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, 24346 Grulleros, Spain
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, 24007 León, Spain
| | - Laura González Del Palacio
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, 24346 Grulleros, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, 24007 León, Spain
| | - F Javier Giráldez
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, 24346 Grulleros, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, 24007 León, Spain
| | - María Martínez-Valladares
- Instituto de Ganadería de Montaña, CSIC-Universidad de León, 24346 Grulleros, Spain
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana, 24007 León, Spain
| |
Collapse
|
40
|
Tamura H. IgA nephropathy associated with Crohn's disease. World J Methodol 2023; 13:67-78. [PMID: 37456980 PMCID: PMC10348078 DOI: 10.5662/wjm.v13.i3.67] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/16/2023] [Accepted: 05/12/2023] [Indexed: 06/20/2023] Open
Abstract
The relationship between IgA nephropathy (IgAN) and Crohn’s disease was reported. IgAN is the most common primary glomerulonephritis and one of the leading causes of chronic kidney disease and end-stage renal failure, and up to 50% of cases progressed to end-stage renal disease within 25 years after IgAN diagnosis. However, specific and effective therapeutic strategies are still lacking. In this review, we discuss the possibility of the mechanism involved in IgAN associated with Crohn’s disease based on the findings of basic and clinical studies. Although the etiology of IgAN associated with Crohn’s disease is not permanent and various factors are thought to be involved, the stabilization of the disease condition of Crohn’s disease is believed to help treat IgAN.
Collapse
Affiliation(s)
- Hiroshi Tamura
- Department of Pediatrics, Kumamoto University, Kumamoto 8608556, Japan
| |
Collapse
|
41
|
Matsumoto K, Sawano H, Otsubo M, Yui A. Comparison of the effects of three forms of soluble dietary fiber on the production of IgA in BALB/cAJcl and BALB/cAJcl-nu/nu mice. J Nutr 2023; 153:1618-1626. [PMID: 37011879 DOI: 10.1016/j.tjnut.2023.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Their ability to induce the production of IgA, especially in the intestine, is one of the health benefits of soluble dietary fibers (SDFs), but the mechanism involved is unclear. OBJECTIVE This study was designed to identify the relationship between the induction of IgA by SDFs and the cecal short-chain fatty acids (SCFAs) content, and to evaluate the importance of T cell-independent IgA production for SDF-induced IgA production. METHODS We compared the SDFs fructooligosaccharides (FO), indigestible glucan (IG), and polydextrose (PD). Male BALB/cAJcl mice or T cell-deficient BALB/cAJcl-nu/nu (nude) mice were fed diets supplemented with one SDF (3% w/w) for 10 weeks and we measured IgA content in their feces, plasma, lung, and submandibular gland. RESULTS In BALB/cAJcl mice, the consumption of all three SDF diets induced fecal IgA production, but the response was stronger in the IG and PD groups than in the FO group. The IgA concentration of the plasma and lung were also higher in the FO and PD groups, and these groups showed significantly higher cecal acetic and n-butyric acid content. In contrast, in nude mice, the induction of IgA production was identified only in fecal samples of mice fed the three SDF diets, even though there were significant increases in cecal SCFAs content. CONCLUSIONS The induction of IgA production by SDFs is occurred T cell-independently in the intestine, but that in the plasma, lung, and submandibular gland is T cell-dependent. SCFAs generated in the large intestine might influence the systemic immune system, but there is no clear relationship between the generation of SCFAs and intestinal IgA production in response to SDF consumption.
Collapse
|
42
|
Vasquez-Martínez N, Guillen D, Moreno-Mendieta SA, Sanchez S, Rodríguez-Sanoja R. The Role of Mucoadhesion and Mucopenetration in the Immune Response Induced by Polymer-Based Mucosal Adjuvants. Polymers (Basel) 2023; 15:1615. [PMID: 37050229 PMCID: PMC10097111 DOI: 10.3390/polym15071615] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Mucus is a viscoelastic gel that acts as a protective barrier for epithelial surfaces. The mucosal vehicles and adjuvants need to pass through the mucus layer to make drugs and vaccine delivery by mucosal routes possible. The mucoadhesion of polymer particle adjuvants significantly increases the contact time between vaccine formulations and the mucosa; then, the particles can penetrate the mucus layer and epithelium to reach mucosa-associated lymphoid tissues. This review presents the key findings that have aided in understanding mucoadhesion and mucopenetration while exploring the influence of physicochemical characteristics on mucus-polymer interactions. We describe polymer-based particles designed with mucoadhesive or mucopenetrating properties and discuss the impact of mucoadhesive polymers on local and systemic immune responses after mucosal immunization. In future research, more attention paid to the design and development of mucosal adjuvants could lead to more effective vaccines.
Collapse
Affiliation(s)
- Nathaly Vasquez-Martínez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
- Programa de Doctorado en Ciencia Bioquímicas, Universidad Nacional Autónoma de México, Circuito de Posgrado, C.U., Coyoacán, Mexico City 04510, Mexico
| | - Daniel Guillen
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
| | - Silvia Andrea Moreno-Mendieta
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
- Programa de Doctorado en Ciencia Bioquímicas, Universidad Nacional Autónoma de México, Circuito de Posgrado, C.U., Coyoacán, Mexico City 04510, Mexico
- Consejo Nacional de Ciencia y Tecnología, Benito Juárez, Mexico City 03940, Mexico
| | - Sergio Sanchez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
| | - Romina Rodríguez-Sanoja
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito, Mario de La Cueva s/n, C.U., Coyoacán, Mexico City 04510, Mexico; (N.V.-M.)
| |
Collapse
|
43
|
Sun Z, Wang X, Feng S, Xie C, Xing Y, Guo L, Zhao J, Ji C. A review of neuroendocrine immune system abnormalities in IBS based on the brain–gut axis and research progress of acupuncture intervention. Front Neurosci 2023; 17:934341. [PMID: 36968497 PMCID: PMC10034060 DOI: 10.3389/fnins.2023.934341] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 02/07/2023] [Indexed: 03/11/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common digestive disorder observed in clinics. Current studies suggest that the pathogenesis of the disease is closely related to abnormal brain–gut interactions, hypokinesia, visceral sensory hypersensitivity in the gastrointestinal tract, and alterations in the intestinal microenvironment. However, it is difficult for a single factor to explain the heterogeneity of symptoms. The Rome IV criteria emphasized the holistic biologic-psycho-social model of IBS, suggesting that symptoms of the disease are closely related to neurogastroenterology and various abnormalities in brain–gut interaction. This study comprehensively reviewed the relationship between the brain–gut axis and IBS, the structure of the brain–gut axis, and the relationship between the brain–gut axis and intestinal microenvironment, and discussed the relationship between the abnormal regulation of the nervous system, endocrine system, and immune system and the incidence of IBS on the basis of brain–gut axis. In terms of treatment, acupuncture therapy can regulate the neuroendocrine-immune system of the body and improve the intestinal microenvironment, and it has the advantages of safety, economy, and effectiveness. We study the pathogenesis of IBS from local to global and micro to macro, and review the use of acupuncture to treat the disease as a whole so as to provide new ideas for the treatment of the disease.
Collapse
Affiliation(s)
- Zhangyin Sun
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Xuejiao Wang
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Shangsheng Feng
- MOE Key Laboratory of Biomedical Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Chaoju Xie
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Yu Xing
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Liang Guo
- College of Acupuncture and Moxibustion, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Jingyu Zhao
- Department of Acupuncture and Moxibustion, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
- *Correspondence: Jingyu Zhao
| | - Changchun Ji
- Department of Acupuncture and Moxibustion, Shaanxi Hospital of Traditional Chinese Medicine, Xi'an, China
- Department of Acupuncture and Moxibustion, Shaanxi Provincial Institute of Traditional Chinese Medicine, Xi'an, China
- Changchun Ji
| |
Collapse
|
44
|
Ruiz-Ojeda FJ, Plaza-Diaz J, Morales J, Álvarez-Calatayud G, Climent E, Silva Á, Martinez-Blanch JF, Enrique M, Tortajada M, Ramon D, Alvarez B, Chenoll E, Gil Á. Effects of a Novel Infant Formula on the Fecal Microbiota in the First Six Months of Life: The INNOVA 2020 Study. Int J Mol Sci 2023; 24:3034. [PMID: 36769356 PMCID: PMC9917896 DOI: 10.3390/ijms24033034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Exclusive breastfeeding is highly recommended for infants for at least the first six months of life. However, for some mothers, it may be difficult or even impossible to do so. This can lead to disturbances in the gut microbiota, which in turn may be related to a higher incidence of acute infectious diseases. Here, we aimed to evaluate whether a novel starting formula versus a standard formula provides a gut microbiota composition more similar to that of breastfed infants in the first 6 months of life. Two hundred and ten infants (70/group) were enrolled in the study and completed the intervention until 12 months of age. For the intervention period, infants were divided into three groups: Group 1 received formula 1 (INN) with a lower amount of protein, a proportion of casein to whey protein ratio of about 70/30 by increasing the content of α-lactalbumin, and with double the amount of docosahexaenoic acid/arachidonic acid than the standard formula; INN also contained a thermally inactivated postbiotic (Bifidobacterium animalis subsp. lactis). Group 2 received the standard formula (STD) and the third group was exclusively breastfed (BF) for exploratory analysis. During the study, visits were made at 21 days, 2, 4, and 6 months of age, with ±3 days for the visit at 21 days of age, ±1 week for the visit at 2 months, and ±2 weeks for the others. Here, we reveal how consuming the INN formula promotes a similar gut microbiota composition to those infants that were breastfed in terms of richness and diversity, genera, such as Bacteroides, Bifidobacterium, Clostridium, and Lactobacillus, and calprotectin and short-chain fatty acid levels at 21 days, 2 and 6 months. Furthermore, we observed that the major bacteria metabolic pathways were more alike between the INN formula and BF groups compared to the STD formula group. Therefore, we assume that consumption of the novel INN formula might improve gut microbiota composition, promoting a healthier intestinal microbiota more similar to that of an infant who receives exclusively human milk.
Collapse
Affiliation(s)
- Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Neuherberg, 85764 Munich, Germany
- Institute of Nutrition and Food Technology “José Mataix”, Centre of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Spain
| | - Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Javier Morales
- Product Development Department, Alter Farmacia SA, 28880 Madrid, Spain
| | | | - Eric Climent
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Ángela Silva
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | | | - María Enrique
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Marta Tortajada
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Daniel Ramon
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Beatriz Alvarez
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Empar Chenoll
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 Paterna, Spain
| | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Centre of Biomedical Research, University of Granada, Avda. del Conocimiento s/n., 18016 Armilla, Spain
- CIBEROBN, CIBER Physiopathology of Obesity and Nutrition, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
45
|
Nakahashi-Ouchida R, Fujihashi K, Kurashima Y, Yuki Y, Kiyono H. Nasal vaccines: solutions for respiratory infectious diseases. Trends Mol Med 2023; 29:124-140. [PMID: 36435633 DOI: 10.1016/j.molmed.2022.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/07/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022]
Abstract
Nasal vaccines induce pathogen-specific dual protective immunity at mucosal surfaces and systemically throughout the body. Consequently, nasal vaccines both prevent pathogen invasion and reduce disease severity. Because of these features, nasal vaccines are considered to be a next-generation tool for preventing respiratory infectious diseases, including COVID-19. However, nasal vaccines must overcome key safety concerns given the anatomic proximity of the central nervous system (CNS) via the olfactory bulbs which lie next to the nasal cavity. This review summarizes current efforts to develop safe and effective nasal vaccines and delivery systems, as well as their clinical applications for the prevention of respiratory infections. We also discuss various concerns regarding the safety of nasal vaccines and introduce a system for evaluating them.
Collapse
Affiliation(s)
- Rika Nakahashi-Ouchida
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, 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
| | - Yosuke Kurashima
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, 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 Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, Chiba, Japan; Chiba University-University of California San Diego (CU-UCSD) Center for Mucosal Immunology, Allergy, and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
| | - Yoshikazu Yuki
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; HanaVax Inc., Tokyo, Japan
| | - Hiroshi Kiyono
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, Chiba, Japan; Chiba University-University of California San Diego (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
|
46
|
Kolenko YG, Timokhina TO, Lynovytska OV, Cherkasova OV, Semenova IS. INDICATORS OF DENTAL HEALTH AND LOCAL IMMUNITY IN YOUNG ADULTS WHO HAVE SUFFERED FROM CORONAVIRUS INFECTION. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2023; 76:1443-1449. [PMID: 37463380 DOI: 10.36740/wlek202306117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
OBJECTIVE The aim: To determine the dental status and state of local immunity in young adults who have suffered from the coronavirus disease. PATIENTS AND METHODS Materials and methods: The main group consisted of 30 people aged 20-22 years, who suffered from the coronavirus infection Covid19 6.1±1.2 months ago. The comparison group included 20 people who did not have a coronavirus infection. The control group consisted of 35 people, randomized by age and sex, who did not have signs of caries and periodontal tissue disease and did not have coronavirus disease. All patients were examined for dental status and local immunity. RESULTS Results: The analysis of indicators of dental status revealed the possibility of the existence of a relationship between the signs of acute SARS-Cov2 viral infection and the development of caries and periodontal tissue diseases. Significant changes in the local immunity of the oral cavity were found in the examined patients, which had a pathogenetic influence on the development and progression of caries and periodontal tissue diseases: a significant increase in the level of Ig G, as well as a probable decrease in the concentration of SIg A relative to the comparison group, a probably higher normative value of pathogenic small- and medium-molecular CICs with a significant decrease in the level of physiological large-sized CICs relative to the comparison group, a decrease in the content of anti-inflammatory IL-4, as well as increased concentration of pro-inflammatory cytokines. CONCLUSION Conclusions: Young adults who have suffered a coronavirus infection during the last 6 months have significantly higher caries prevalence, bleeding index, PMA index and hygiene index, halitosis, which indicates deeper tissue damage and tooth pathology with the formation of dentition defects than in the comparison group. Indicators of local immunity of the oral fluid have a deep and specific character.
Collapse
|
47
|
Li X, Li Z, Zhang X, Zeng Q, Huang X, Sheng L, Ahn DU, Cai Z. Restoration of immunity by whole egg was superior to egg white or egg yolk in a cyclophosphamide-induced immunocompromised mouse model. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
48
|
Prioritizing autoimmunity risk variants for functional analyses by fine-mapping mutations under natural selection. Nat Commun 2022; 13:7069. [PMID: 36400766 PMCID: PMC9674589 DOI: 10.1038/s41467-022-34461-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 10/25/2022] [Indexed: 11/19/2022] Open
Abstract
Pathogen-driven selection shaped adaptive mutations in immunity genes, including those contributing to inflammatory disorders. Functional characterization of such adaptive variants can shed light on disease biology and past adaptations. This popular idea, however, was difficult to test due to challenges in pinpointing adaptive mutations in selection footprints. In this study, using a local-tree-based approach, we show that 28% of risk loci (153/535) in 21 inflammatory disorders bear footprints of moderate and weak selection, and part of them are population specific. Weak selection footprints allow partial fine-mapping, and we show that in 19% (29/153) of the risk loci under selection, candidate disease variants are hitchhikers, and only in 39% of cases they are likely selection targets. We predict function for a subset of these selected SNPs and highlight examples of antagonistic pleiotropy. We conclude by offering disease variants under selection that can be tested functionally using infectious agents and other stressors to decipher the poorly understood link between environmental stressors and genetic risk in inflammatory conditions.
Collapse
|
49
|
Nakanishi K, Takase T, Ohira Y, Ida R, Mogi N, Kikuchi Y, Matsuda M, Kurohane K, Akimoto Y, Hayakawa J, Kawakami H, Niwa Y, Kobayashi H, Umemoto E, Imai Y. Prevention of Shiga toxin 1-caused colon injury by plant-derived recombinant IgA. Sci Rep 2022; 12:17999. [PMID: 36289440 PMCID: PMC9606113 DOI: 10.1038/s41598-022-22851-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023] Open
Abstract
Immunoglobulin A (IgA) is a candidate antibody for oral passive immunization against mucosal pathogens like Shiga toxin-producing Escherichia coli (STEC). We previously established a mouse IgG monoclonal antibody (mAb) neutralizing Shiga toxin 1 (Stx1), a bacterial toxin secreted by STEC. We designed cDNA encoding an anti-Stx1 antibody, in which variable regions were from the IgG mAb and all domains of the heavy chain constant region from a mouse IgA mAb. Considering oral administration, we expressed the cDNA in a plant expression system aiming at the production of enough IgA at low cost. The recombinant-IgA expressed in Arabidopsis thaliana formed the dimeric IgA, bound to the B subunit of Stx1, and neutralized Stx1 toxicity to Vero cells. Colon injury was examined by exposing BALB/c mice to Stx1 via the intrarectal route. Epithelial cell death, loss of crypt and goblet cells from the distal colon were observed by electron microscopy. A loss of secretory granules containing MUC2 mucin and activation of caspase-3 were observed by immunohistochemical methods. Pretreatment of Stx1 with the plant-based recombinant IgA completely suppressed caspase-3 activation and loss of secretory granules. The results indicate that a plant-based recombinant IgA prevented colon damage caused by Stx1 in vivo.
Collapse
Affiliation(s)
- Katsuhiro Nakanishi
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Taichi Takase
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Yuya Ohira
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Ryota Ida
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Noriko Mogi
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Yuki Kikuchi
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Minami Matsuda
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Kohta Kurohane
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Yoshihiro Akimoto
- grid.411205.30000 0000 9340 2869Department of Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611 Japan
| | - Junri Hayakawa
- grid.411205.30000 0000 9340 2869Laboratory for Electron Microscopy, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611 Japan
| | - Hayato Kawakami
- grid.411205.30000 0000 9340 2869Department of Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo, 181-8611 Japan
| | - Yasuo Niwa
- grid.469280.10000 0000 9209 9298Laboratory of Plant Molecular Improvement, Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Hirokazu Kobayashi
- grid.469280.10000 0000 9209 9298Laboratory of Plant Molecular Improvement, Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Eiji Umemoto
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| | - Yasuyuki Imai
- grid.469280.10000 0000 9209 9298Laboratory of Microbiology and Immunology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka City, Shizuoka 422-8526 Japan
| |
Collapse
|
50
|
Madhavan M, Ritchie AJ, Aboagye J, Jenkin D, Provstgaad-Morys S, Tarbet I, Woods D, Davies S, Baker M, Platt A, Flaxman A, Smith H, Belij-Rammerstorfer S, Wilkins D, Kelly EJ, Villafana T, Green JA, Poulton I, Lambe T, Hill AVS, Ewer KJ, Douglas AD. Tolerability and immunogenicity of an intranasally-administered adenovirus-vectored COVID-19 vaccine: An open-label partially-randomised ascending dose phase I trial. EBioMedicine 2022; 85:104298. [PMID: 36229342 PMCID: PMC9550199 DOI: 10.1016/j.ebiom.2022.104298] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Intranasal vaccination may induce protective local and systemic immune responses against respiratory pathogens. A number of intranasal SARS-CoV-2 vaccine candidates have achieved protection in pre-clinical challenge models, including ChAdOx1 nCoV-19 (AZD1222, University of Oxford / AstraZeneca). METHODS We performed a single-centre open-label Phase I clinical trial of intranasal vaccination with ChAdOx1 nCoV-19 in healthy adults, using the existing formulation produced for intramuscular administration. Thirty SARS-CoV-2 vaccine-naïve participants were allocated to receive 5 × 109 viral particles (VP, n=6), 2 × 1010 VP (n=12), or 5 × 1010 VP (n=12). Fourteen received second intranasal doses 28 days later. A further 12 received non-study intramuscular mRNA SARS-CoV-2 vaccination between study days 22 and 46. To investigate intranasal ChAdOx1 nCoV-19 as a booster, six participants who had previously received two intramuscular doses of ChAdOx1 nCoV-19 and six who had received two intramuscular doses of BNT162b2 (Pfizer / BioNTech) were given a single intranasal dose of 5 × 1010 VP of ChAdOx1 nCoV-19. Objectives were to assess safety (primary) and mucosal antibody responses (secondary). FINDINGS Reactogenicity was mild or moderate. Antigen-specific mucosal antibody responses to intranasal vaccination were detectable in a minority of participants, rarely exceeding levels seen after SARS-CoV-2 infection. Systemic responses to intranasal vaccination were typically weaker than after intramuscular vaccination with ChAdOx1 nCoV-19. Antigen-specific mucosal antibody was detectable in participants who received an intramuscular mRNA vaccine after intranasal vaccination. Seven participants developed symptomatic SARS-CoV-2 infection. INTERPRETATION This formulation of intranasal ChAdOx1 nCoV-19 showed an acceptable tolerability profile but induced neither a consistent mucosal antibody response nor a strong systemic response. FUNDING AstraZeneca.
Collapse
Affiliation(s)
- Meera Madhavan
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK; Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Adam J Ritchie
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Jeremy Aboagye
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Daniel Jenkin
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK; Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Samuel Provstgaad-Morys
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Iona Tarbet
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Danielle Woods
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Sophie Davies
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Megan Baker
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Abigail Platt
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Amy Flaxman
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Holly Smith
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | | | - Deidre Wilkins
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, 1 Medimmune Way, Gaithersburg, MD 20878, USA
| | - Elizabeth J Kelly
- Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, 1 Medimmune Way, Gaithersburg, MD 20878, USA
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Justin A Green
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ian Poulton
- Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Teresa Lambe
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK; Oxford Vaccine Group, Centre for Clinical Vaccinology and Tropical Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK; China Academy of Medical Sciences Oxford Institute, University of Oxford, NDM Research Building, Old Road Campus, Headington, Oxford OX3 7FZ, UK
| | - Adrian V S Hill
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Katie J Ewer
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK
| | - Alexander D Douglas
- Jenner Institute, University of Oxford, Old Road Campus Research Building, Oxford OX3 7BN, UK.
| |
Collapse
|