1
|
Surd AO, Răducu C, Răducu E, Ihuț A, Munteanu C. Lamina Propria and GALT: Their Relationship with Different Gastrointestinal Diseases, Including Cancer. GASTROINTESTINAL DISORDERS 2024; 6:947-963. [DOI: 10.3390/gidisord6040066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
Abstract
The structural integrity of the gastrointestinal tract is important because it dictates the functionality of this system. Regarding this, gut-associated lymphoid tissue (GALT) has a significant role in immunity. Most cancer research focuses on organized lymphoid structures and less on diffuse structures such as the lamina propria (LP). Therefore, this paper aims to investigate the link between the LP and cancer in humans. The interstitial matrix and loose connective tissue layer located directly under the epithelium is known as the LP. In this area, there are a lot of IgA+ plasma cells (PCs), T and B lymphocytes, macrophages, dendritic cells (DCs), and stromal cells (SCs). Antigens from the lumen are picked up by LP DCs and presented directly to B cells, which may cause IgA class switching and differentiation in the presence of T cells. In humans, the GALT of the mucosa has been proposed as the source of a unique malignancy known as “GALT carcinoma”, which is thought to represent the “third pathway of colorectal carcinogenesis”. However, present colorectal cancer classifications do not define GALT carcinoma as a separate histologic category.
Collapse
Affiliation(s)
- Adrian Onisim Surd
- Department of Pediatric Surgery, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Camelia Răducu
- Department of Technological Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania
| | - Eugen Răducu
- Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Andrada Ihuț
- Department of Technological Sciences, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania
| | - Camelia Munteanu
- Department of Plant Culture, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania
| |
Collapse
|
2
|
Reddiar SB, Xie Y, Abdallah M, Han S, Hu L, Feeney OM, Gracia G, Anshabo A, Lu Z, Farooq MA, Styles IK, Phillips ARJ, Windsor JA, Porter CJH, Cao E, Trevaskis NL. Intestinal Lymphatic Biology, Drug Delivery, and Therapeutics: Current Status and Future Directions. Pharmacol Rev 2024; 76:1326-1398. [PMID: 39179383 DOI: 10.1124/pharmrev.123.001159] [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: 12/19/2023] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Historically, the intestinal lymphatics were considered passive conduits for fluids, immune cells, dietary lipids, lipid soluble vitamins, and lipophilic drugs. Studies of intestinal lymphatic drug delivery in the late 20th century focused primarily on the drugs' physicochemical properties, especially high lipophilicity, that resulted in intestinal lymphatic transport. More recent discoveries have changed our traditional view by demonstrating that the lymphatics are active, plastic, and tissue-specific players in a range of biological and pathological processes, including within the intestine. These findings have, in turn, inspired exploration of lymph-specific therapies for a range of diseases, as well as the development of more sophisticated strategies to actively deliver drugs or vaccines to the intestinal lymph, including a range of nanotechnologies, lipid prodrugs, and lipid-conjugated materials that "hitchhike" onto lymphatic transport pathways. With the increasing development of novel therapeutics such as biologics, there has been interest in whether these therapeutics are absorbed and transported through intestinal lymph after oral administration. Here we review the current state of understanding of the anatomy and physiology of the gastrointestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. We summarize the current state-of-the-art approaches to deliver drugs and quantify their uptake into the intestinal lymphatic system. Finally, and excitingly, we discuss recent examples of significant pharmacokinetic and therapeutic benefits achieved via intestinal lymphatic drug delivery. We also propose approaches to advance the development and clinical application of intestinal lymphatic delivery strategies in the future. SIGNIFICANCE STATEMENT: This comprehensive review details the understanding of the anatomy and physiology of the intestinal lymphatic system in health and disease, with a focus on aspects relevant to drug delivery. It highlights current state-of-the-art approaches to deliver drugs to the intestinal lymphatics and the shift toward the use of these strategies to achieve pharmacokinetic and therapeutic benefits for patients.
Collapse
Affiliation(s)
- Sanjeevini Babu Reddiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Yining Xie
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Mohammad Abdallah
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Sifei Han
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Luojuan Hu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Orlagh M Feeney
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Gracia Gracia
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Abel Anshabo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Zijun Lu
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Muhammad Asim Farooq
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Ian K Styles
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Anthony R J Phillips
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - John A Windsor
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Christopher J H Porter
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Enyuan Cao
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| | - Natalie L Trevaskis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia (S.B.R., Y.X., M.A., S.H., L.H., O.M.F., G.G., A.A., Z.L., M.A.F., I.K.S., C.J.H.P., E.C., N.L.T.); China Pharmaceutical University, Nanjing, China (S.H., L.H.); Applied Surgery and Metabolism Laboratory, School of Biological Sciences (A.R.J.P.) and Surgical and Translational Research Centre, Department of Surgery, Faculty of Medical and Health Sciences (A.R.J.P., J.A.W.), University of Auckland, Auckland, New Zealand; and Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (N.L.T.)
| |
Collapse
|
3
|
Ding M, Li B, Chen H, Ross RP, Stanton C, Zhao J, Chen W, Yang B. Bifidobacterium longum Subsp. infantis Promotes IgA Level of Growing Mice in a Strain-Specific and Intestinal Niche-Dependent Manner. Nutrients 2024; 16:1148. [PMID: 38674840 PMCID: PMC11054607 DOI: 10.3390/nu16081148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Throughout infancy, IgA is crucial for maintaining gut mucosal immunity. This study aims to determine whether supplementing newborn mice with eight different strains of Bifidobacterium longum subsp. infantis might regulate their IgA levels. The strains were gavaged to BALB/C female (n = 8) and male (n = 8) dams at 1-3 weeks old. Eight strains of B. longum subsp. infantis had strain-specific effects in the regulation of intestinal mucosal barriers. B6MNI, I4MI, and I10TI can increase the colonic IgA level in females and males. I8TI can increase the colonic IgA level in males. B6MNI was also able to significantly increase the colonic sIgA level in females. B6MNI, I4MI, I8TI, and I10TI regulated colonic and Peyer's patch IgA synthesis genes but had no significant effect on IgA synthesis pathway genes in the jejunum and ileum. Moreover, the variety of sIgA-coated bacteria in male mice was changed by I4MI, I5TI, I8TI, and B6MNI. These strains also can decrease the relative abundance of Escherichia coli. These results indicate that B. longum subsp. infantis can promote IgA levels but show strain specificity. Different dietary habits with different strains of Bifidobacterium may have varying effects on IgA levels when supplemented in early infancy.
Collapse
Affiliation(s)
- Mengfan Ding
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bowen Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Reynolds Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
- APC Microbiome Ireland, University College Cork, T12 R229 Cork, Ireland
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
- APC Microbiome Ireland, University College Cork, T12 R229 Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, P61 C996 Cork, Ireland
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; (M.D.); (B.L.); (H.C.); (J.Z.); (W.C.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China; (R.P.R.); (C.S.)
| |
Collapse
|
4
|
Liu X, Wang X, Zhang P, Fang Y, Liu Y, Ding Y, Zhang W. Intestinal homeostasis in the gut-lung-kidney axis: a prospective therapeutic target in immune-related chronic kidney diseases. Front Immunol 2023; 14:1266792. [PMID: 38022571 PMCID: PMC10646503 DOI: 10.3389/fimmu.2023.1266792] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
In recent years, the role of intestinal homeostasis in health has received increasing interest, significantly improving our understanding of the complex pathophysiological interactions of the gut with other organs. Microbiota dysbiosis, impaired intestinal barrier, and aberrant intestinal immunity appear to contribute to the pathogenesis of immune-related chronic kidney diseases (CKD). Meanwhile, the relationship between the pathological changes in the respiratory tract (e.g., infection, fibrosis, granuloma) and immune-related CKD cannot be ignored. The present review aimed to elucidate the new underlying mechanism of immune-related CKD. The lungs may affect kidney function through intestinal mediation. Communication is believed to exist between the gut and lung microbiota across long physiological distances. Following the inhalation of various pathogenic factors (e.g., particulate matter 2.5 mum or less in diameter, pathogen) in the air through the mouth and nose, considering the anatomical connection between the nasopharynx and lungs, gut microbiome regulates oxidative stress and inflammatory states in the lungs and kidneys. Meanwhile, the intestine participates in the differentiation of T cells and promotes the migration of various immune cells to specific organs. This better explain the occurrence and progression of CKD caused by upper respiratory tract precursor infection and suggests the relationship between the lungs and kidney complications in some autoimmune diseases (e.g., anti-neutrophil cytoplasm antibodies -associated vasculitis, systemic lupus erythematosus). CKD can also affect the progression of lung diseases (e.g., acute respiratory distress syndrome and chronic obstructive pulmonary disease). We conclude that damage to the gut barrier appears to contribute to the development of immune-related CKD through gut-lung-kidney interplay, leading us to establish the gut-lung-kidney axis hypothesis. Further, we discuss possible therapeutic interventions and targets. For example, using prebiotics, probiotics, and laxatives (e.g., Rhubarb officinale) to regulate the gut ecology to alleviate oxidative stress, as well as improve the local immune system of the intestine and immune communication with the lungs and kidneys.
Collapse
Affiliation(s)
- Xinyin Liu
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- Department of Traditional Chinese Medicine, Jiande First People’s Hospital, Jiande, Hangzhou, China
| | - Xiaoran Wang
- Department of Nephrology, The First People’s Hospital of Hangzhou Lin’an District, Hangzhou, China
| | - Peipei Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yiwen Fang
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanyan Liu
- Department of Geriatric, Zhejiang Aged Care Hospital, Hangzhou, China
| | - Yueyue Ding
- Department of Geriatric, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Wen Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
5
|
Bamias G, Kitsou K, Rivera-Nieves J. The Underappreciated Role of Secretory IgA in IBD. Inflamm Bowel Dis 2023; 29:1327-1341. [PMID: 36943800 PMCID: PMC10393212 DOI: 10.1093/ibd/izad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Indexed: 03/23/2023]
Abstract
Eighty percent of antibody secreting cells (ASCs) are found in the intestine, where they produce grams of immunoglobulin (Ig) A daily. immunoglobulin A is actively transcytosed into the lumen, where it plays a critical role in modulating the gut microbiota. Although loss of immune tolerance to bacterial antigens is the likely trigger of the dysregulated immune response that characterizes inflammatory bowel disease (IBD), little effort has been placed on understanding the interface between B cells, IgA, and the microbiota during initiation or progression of disease. This may be in part due to the misleading fact that IgA-deficient humans are mostly asymptomatic, likely due to redundant role of secretory (S) IgM. Intestinal B cell recruitment is critically dependent on integrin α4β7-MAdCAM-1 interactions, yet antibodies that target α4β7 (ie, vedolizumab), MAdCAM-1 (ie, ontamalimab), or both β7 integrins (α4β7 and αE [CD103] β7; etrolizumab) are in clinical use or development as IBD therapeutics. The effect of such interventions on the biology of IgA is largely unknown, yet a single dose of vedolizumab lowers SIgA levels in stool and weakens the oral immunization response to cholera vaccine in healthy volunteers. Thus, it is critical to further understand the role of these integrins for the migration of ASC and other cellular subsets during homeostasis and IBD-associated inflammation and the mode of action of drugs that interfere with this traffic. We have recently identified a subset of mature ASC that employs integrin αEβ7 to dock with intestinal epithelial cells, predominantly in the pericryptal region of the terminal ileum. This role for the integrin had not been appreciated previously, nor the αEβ7-dependent mechanism of IgA transcytosis that it supports. Furthermore, we find that B cells more than T cells are critically dependent on α4β7-MAdCAM-1 interactions; thus MAdCAM-1 blockade and integrin-β7 deficiency counterintuitively hasten colitis in interleukin-10-deficient mice. In both cases, de novo recruitment of IgA ASC to the intestinal lamina propria is compromised, leading to bacterial overgrowth, dysbiosis, and lethal colitis. Thus, despite the safe and effective use of anti-integrin antibodies in patients with IBD, much remains to be learned about their various cell targets.
Collapse
Affiliation(s)
- Giorgos Bamias
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Konstantina Kitsou
- GI Unit, 3rd Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Sotiria Hospital, Athens, Greece
| | - Jesús Rivera-Nieves
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA, USA
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
Donovan SM, Abrams SA, Azad MB, Belfort MB, Bode L, Carlson SE, Dallas DC, Hettinga K, Järvinen K, Kim JH, Lebrilla CB, McGuire MK, Sela DA, Neu J. Summary of the joint National Institutes of Health and the Food and Drug Administration workshop titled "exploring the science surrounding the safe use of bioactive ingredients in infant formula: Considerations for an assessment framework". J Pediatr 2022; 255:30-41.e1. [PMID: 36463938 PMCID: PMC10121942 DOI: 10.1016/j.jpeds.2022.11.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/20/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022]
Affiliation(s)
- Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL
| | - Steven A Abrams
- Department of Pediatrics Dell Medical School, The University of Texas at Austin, Austin, TX
| | - Meghan B Azad
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Canada; Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba, Winnipeg, Canada
| | - Mandy B Belfort
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lars Bode
- Department of Pediatrics and Mother-Milk-Infant Center of Research Excellence (MOMI CORE), University of California, San Diego, La Jolla, CA
| | - Susan E Carlson
- Department of Dietetics and Nutrition, Kansas University Medical Center and The University of Kansas, Kansas City, KS
| | - David C Dallas
- Department of Nutrition, Oregon State University, Corvallis, OR
| | - Kasper Hettinga
- Department of Food Sciences and Agrotechnology, Wageningen University, Wageningen, Netherlands
| | - Kirsi Järvinen
- Department of Pediatrics, Golisano Children's Hospital and University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Jae H Kim
- Perinatal Institute, Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH
| | | | | | - David A Sela
- Department of Food Science, University of Massachusetts, Amherst, Amherst, MA
| | - Josef Neu
- Department of Pediatrics, University of Florida, Gainesville, FL.
| |
Collapse
|
7
|
Zhu Y, Zhao Q, Huang Q, Li Y, Yu J, Zhang R, Liu J, Yan P, Xia J, Guo L, Liu G, Yang X, Zeng J. Nuciferine Regulates Immune Function and Gut Microbiota in DSS-Induced Ulcerative Colitis. Front Vet Sci 2022; 9:939377. [PMID: 35909691 PMCID: PMC9328756 DOI: 10.3389/fvets.2022.939377] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Nuciferine, a major aporphine alkaloid obtained from the leaves of Nelumbo nucifera, exhibits anti-cancer and anti-inflammatory properties; however, its protective effects against inflammatory bowel diseases (IBD) has never been explored. In this study, an ulcerative colitis (UC) model was established in BALb/c mice by the continuous administration of 5% dextran sulfate sodium (DSS) in drinking water for 1 week. From day 8 to day 14, the DSS-treated mice were divided into a high-dose and a low-dose nuciferine treatment group and were intraperitoneally injected with the corresponding dose of the drug. Body weight loss, disease activity index (DAI), and colon length were measured. Histological changes were observed using hematoxylin and eosin staining. T lymphocyte proliferation was assessed by MTT assay. The ratio of CD3+, CD4+, CD8+, Th1, Th2, Th17, and Treg cells were estimated by flow cytometry. Finally, 16S rRNA sequencing was performed to compare the composition and relative abundance of the gut microbiota among the different treatment groups. The results showed that nuciferine treatment led to a significant improvement in symptoms, such as histological injury and colon shortening in mice with DSS-induced UC. Nuciferine treatment improved the Th1/Th2 and Treg/Th17 balance in the DSS-induced IBD model, as well as the composition of the intestinal microflora. At the phylum level, compared with the control group, the abundance of Firmicutes and Actinobacteriota was decreased in the model group, whereas that of Bacteroidetes increased. Meanwhile, at the genus level, compared with the control group, the numbers of the genera Lachnospiraceae_Clostridium, Bilophila and Halomonas reduced in the model group, while those of Bacteroides, Parabacteroides, and Paraprevotella increased. Notably, nuciferine administration reversed this DSS-induced gut dysbiosis. These results indicated that nuciferine modulates gut microbiota homeostasis and immune function in mice with DSS-induced UC.
Collapse
Affiliation(s)
- Yiling Zhu
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Qing Zhao
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Qi Huang
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Yana Li
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Jie Yu
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Rui Zhang
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Jiali Liu
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Pupu Yan
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Jinjin Xia
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Liwei Guo
- College of Animal Science, Yangtze University, Jingzhou, China
- *Correspondence: Liwei Guo
| | - Guoping Liu
- College of Animal Science, Yangtze University, Jingzhou, China
- Guoping Liu
| | - Xiaolin Yang
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Jianguo Zeng
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| |
Collapse
|
8
|
Bos A, van Egmond M, Mebius R. The role of retinoic acid in the production of immunoglobulin A. Mucosal Immunol 2022; 15:562-572. [PMID: 35418672 DOI: 10.1038/s41385-022-00509-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/09/2022] [Accepted: 03/26/2022] [Indexed: 02/04/2023]
Abstract
Vitamin A and its derivative retinoic acid (RA) play important roles in the regulation of mucosal immunity. The effect of vitamin A metabolism on T lymphocyte immunity has been well documented, but its role in mucosal B lymphocyte regulation is less well described. Intestinal immunoglobulin A (IgA) is key in orchestrating a balanced gut microbiota composition. Here, we describe the contribution of RA to IgA class switching in tissues including the lamina propria, mesenteric lymph nodes, Peyer's patches and isolated lymphoid follicles. RA can either indirectly skew T cells or directly affect B cell differentiation. IgA levels in healthy individuals are under the control of the metabolism of vitamin A, providing a steady supply of RA. However, IgA levels are altered in inflammatory bowel disease patients, making control of the metabolism of vitamin A a potential therapeutic target. Thus, dietary vitamin A is a key player in regulating IgA production within the intestine, acting via multiple immunological pathways.
Collapse
Affiliation(s)
- Amelie Bos
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Research Institute of Amsterdam Institute for Infection and Immunity, Vrije Universiteit, Amsterdam, The Netherlands
| | - Marjolein van Egmond
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Research Institute of Amsterdam Institute for Infection and Immunity, Vrije Universiteit, Amsterdam, The Netherlands.,Amsterdam UMC, Department of Surgery, Research Institute of Amsterdam Institute for Infection and Immunity, Vrije Universiteit, Amsterdam, The Netherlands
| | - Reina Mebius
- Amsterdam UMC, Department of Molecular Cell Biology and Immunology, Research Institute of Amsterdam Institute for Infection and Immunity, Vrije Universiteit, Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Liu N, Feng G, Zhang X, Hu Q, Sun S, Sun J, Sun Y, Wang R, Zhang Y, Wang P, Li Y. The Functional Role of Lactoferrin in Intestine Mucosal Immune System and Inflammatory Bowel Disease. Front Nutr 2021; 8:759507. [PMID: 34901112 PMCID: PMC8655231 DOI: 10.3389/fnut.2021.759507] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/18/2021] [Indexed: 12/21/2022] Open
Abstract
Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD), is one of the main types of intestinal inflammatory diseases with intestine mucosal immune disorder. Intestine mucosal immune system plays a remarkable and important role in the etiology and pathogenesis of IBD. Therefore, understanding the intestine mucosal immune mechanism is a key step to develop therapeutic interventions for IBD. Intestine mucosal immune system and IBD are influenced by various factors, such as inflammation, gut permeability, gut microbiota, and nutrients. Among these factors, emerging evidence show that nutrients play a key role in inflammation activation, integrity of intestinal barrier, and immune cell modulation. Lactoferrin (LF), an iron-binding glycoprotein belonging to transferrin family, is a dietary bioactive component abundantly found in mammalian milk. Notably, LF has been reported to perform diverse biological functions including antibacterial activity, anti-inflammatory activity, intestinal barrier protection, and immune cell modulation, and is involved in maintaining intestine mucosal immune homeostasis. The improved understanding of the properties of LF in intestine mucosal immune system and IBD will facilitate its application in nutrition, clinical medicine, and health. Herein, this review outlines the recent advancements on LF as a potential therapeutic intervention for IBD associated with intestine mucosal immune system dysfunction. We hope this review will provide a reference for future studies and lay a theoretical foundation for LF-based therapeutic interventions for IBD by understanding the particular effects of LF on intestine mucosal immune system.
Collapse
Affiliation(s)
- Ning Liu
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Gang Feng
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- Yili Maternal & Infant Nutrition Institute, Beijing, China
| | - Xiaoying Zhang
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- Yili Maternal & Infant Nutrition Institute, Beijing, China
| | - Qingjuan Hu
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Shiqiang Sun
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Jiaqi Sun
- Inner Mongolia Yili Industrial Group, Co., Ltd., Hohhot, China
- Yili Maternal & Infant Nutrition Institute, Beijing, China
| | - Yanan Sun
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yan Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- College of Food Science and Engineering, Gansu Agricultural University, Lanzhou, China
| | - Pengjie Wang
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
- Key Laboratory of Functional Dairy, Ministry of Education, Department of Nutrition and Health, China Agricultural University, Beijing, China
| |
Collapse
|
10
|
Abstract
IgA mediates microbial homeostasis at the intestinal mucosa. Within the gut, IgA acts in a context-dependent manner to both prevent and promote bacterial colonization and to influence bacterial gene expression, thus providing exquisite control of the microbiota. IgA-microbiota interactions are highly diverse across individuals and populations, yet the factors driving this variation remain poorly understood. In this Review, we summarize evidence for the host, bacterial and environmental factors that influence IgA-microbiota interactions. Recent advances have helped to clarify the antigenic specificity and immune selection of intestinal IgA and have highlighted the importance of microbial glycan recognition. Furthermore, emerging evidence suggests that diet and nutrition play an important role in shaping IgA recognition of the microbiota. IgA-microbiota interactions are disrupted during both overnutrition and undernutrition and may be altered dynamically in response to diet, with potential implications for host health. We situate this research in the context of outstanding questions and future directions in order to better understand the fascinating paradigm of IgA-microbiota homeostasis.
Collapse
|
11
|
Taylor RA, Xiao S, Carias AM, McRaven MD, Thakkar DN, Araínga M, Allen EJ, Rogers KA, Kumarapperuma SC, Gong S, Fought AJ, Anderson MR, Thomas Y, Schneider JR, Goins B, Fox P, Villinger FJ, Ruprecht RM, Hope TJ. PET/CT targeted tissue sampling reveals virus specific dIgA can alter the distribution and localization of HIV after rectal exposure. PLoS Pathog 2021; 17:e1009632. [PMID: 34061907 PMCID: PMC8195437 DOI: 10.1371/journal.ppat.1009632] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/11/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022] Open
Abstract
Human immunodeficiency virus (HIV) vaccines have not been successful in clinical trials. Dimeric IgA (dIgA) in the form of secretory IgA is the most abundant antibody class in mucosal tissues, making dIgA a prime candidate for potential HIV vaccines. We coupled Positron Emission Tomography (PET) imaging and fluorescent microscopy of 64Cu-labeled, photoactivatable-GFP HIV (PA-GFP-BaL) and fluorescently labeled dIgA to determine how dIgA antibodies influence virus interaction with mucosal barriers and viral penetration in colorectal tissue. Our results show that HIV virions rapidly disseminate throughout the colon two hours after exposure. The presence of dIgA resulted in an increase in virions and penetration depth in the transverse colon. Moreover, virions were found in the mesenteric lymph nodes two hours after viral exposure, and the presence of dIgA led to an increase in virions in mesenteric lymph nodes. Taken together, these technologies enable in vivo and in situ visualization of antibody-virus interactions and detailed investigations of early events in HIV infection.
Collapse
Affiliation(s)
- Roslyn A. Taylor
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Sixia Xiao
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Ann M. Carias
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Michael D. McRaven
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Divya N. Thakkar
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Mariluz Araínga
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
| | - Edward J. Allen
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Kenneth A. Rogers
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
| | - Sidath C. Kumarapperuma
- Research Imaging Institute, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| | - Siqi Gong
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas, United States of America
- Texas Biomedical Research Institute and Southwest National Primate Research Center, San Antonio, Texas, United States of America
| | - Angela J. Fought
- Department of Preventative Medicine, Division of Biostatistics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Meegan R. Anderson
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Yanique Thomas
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Jeffrey R. Schneider
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Beth Goins
- Research Imaging Institute, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| | - Peter Fox
- Research Imaging Institute, University of Texas Health San Antonio, San Antonio, Texas, United States of America
| | - Francois J. Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
| | - Ruth M. Ruprecht
- New Iberia Research Center, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
- Department of Biology, University of Louisiana at Lafayette, Lafayette, Louisiana, United States of America
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas Health San Antonio, San Antonio, Texas, United States of America
- Texas Biomedical Research Institute and Southwest National Primate Research Center, San Antonio, Texas, United States of America
| | - Thomas J. Hope
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
12
|
Simeonova D, Stoyanov D, Leunis JC, Carvalho AF, Kubera M, Murdjeva M, Maes M. Increased Serum Immunoglobulin Responses to Gut Commensal Gram-Negative Bacteria in Unipolar Major Depression and Bipolar Disorder Type 1, Especially When Melancholia Is Present. Neurotox Res 2020; 37:338-348. [PMID: 31802379 DOI: 10.1007/s12640-019-00126-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/28/2019] [Accepted: 10/18/2019] [Indexed: 01/05/2023]
Abstract
Major depressive disorder (MDD) is accompanied by higher serum IgM/IgA responses to LPS of Gram-negative bacteria, suggesting increased bacterial translocation and gut dysbiosis while the latter may occur in bipolar disorder (BD). There are differences between MDD and BD type 1 (BP1) and 2 (BP2) in nitro-oxidative stress biomarkers associated with leaky gut. This study examines serum IgM/IgA responses directed to LPS of 6 Gram-negative bacteria as well as IgG responses to oxidized LDL (oxLDL) in 29 BP1, 37 BP2, 44 MDD, and 30 healthy individuals. Increased IgM/IgA responses to Pseudomonas aeruginosa significantly discriminated patients with affective disorders (MDD plus BD) from controls. BP1 patients showed higher IgM responses to Morganella morganii as compared with MDD and BP2 patients. Patients with melancholia showed higher IgA responses to Citrobacter koseri as compared to controls and non-melancholic depression. The total score on the Hamilton Depression Rating Scale was significantly associated with IgA responses to C. koseri. IgG to oxLDL was significantly associated with increased bacterial translocation. In conclusion, MDD, BP1, and BP2 are accompanied by an immune response due to the increased load of LPS while these aberrations in the gut-brain axis are most pronounced in BP1 and melancholia. Activated oxidative stress pathways and autoimmune responses to oxidative specific epitopes in mood disorders may be driven by a breakdown in gut paracellular, transcellular, and/or vascular pathways. If replicated, drugs that protect the integrity of the gut barrier may offer novel therapeutic opportunities for BP1 and MDD.
Collapse
Affiliation(s)
- Denitsa Simeonova
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Drozdstoy Stoyanov
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria
| | | | - Andre F Carvalho
- Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Marta Kubera
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Marianna Murdjeva
- Department of Microbiology and Immunology, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
- Section of Immunological Assessment of Chronic Stress, Technological Center of Emergency Medicine, Plovdiv, Bulgaria
| | - Michael Maes
- Department of Psychiatry and Medical Psychology, Medical Faculty, Medical University of Plovdiv, Plovdiv, Bulgaria.
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Impact Research Center, Deakin University, Geelong, Australia.
| |
Collapse
|
13
|
Fourcade L, Sabourin-Poirier C, Perraud V, Faucher MC, Chagnon-Choquet J, Labbé AC, Alary M, Guédou F, Poudrier J, Roger M. Natural Immunity to HIV is associated with Low BLyS/BAFF levels and low frequencies of innate marginal zone like CD1c+ B-cells in the genital tract. PLoS Pathog 2019; 15:e1007840. [PMID: 31173604 PMCID: PMC6583986 DOI: 10.1371/journal.ppat.1007840] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/19/2019] [Accepted: 05/14/2019] [Indexed: 12/29/2022] Open
Abstract
BLyS/BAFF is recognized for its role in B-cell ontogenesis, as well as cell fate decision towards the first-line/innate marginal zone (MZ) B-cell pool. Excess BLyS/BAFF is associated with hyperglobulinemia and increased frequencies of activated precursor-like MZ B-cells. Herein, we show that HIV highly-exposed seronegative (HESN) commercial sex workers (CSWs) had lower soluble BLyS/BAFF levels and relative frequencies of BLyS/BAFF expressing cells in their genital mucosa when compared to those from HIV-infected CSWs and HIV-uninfected non-CSWs. Furthermore, we identified genital innate and/or marginal zone (MZ)-like CD1c+ B-cells that naturally bind to fully glycosylated gp120, which frequencies were lower in HESNs when compared to HIV-infected CSWs and HIV-uninfected non-CSWs. Although genital levels of total IgA were similar between groups, HESNs had lower levels of total IgG1 and IgG3. Interestingly, HIV-gp41 reactive IgG1 were found in some HESNs. Low genital levels of BLyS/BAFF observed in HESNs may allow for controlled first-line responses, contributing to natural immunity to HIV. Worldwide, most human immunodeficiency virus (HIV) infections affect women through heterosexual intercourse. We and others have identified African female commercial sex workers (CSWs), who remain seronegative despite high exposition to HIV (HESNs). Innate marginal zone (MZ) B-cells recirculate in humans and have been found in front-line areas such as the sub-epithelial lamina propria of mucosal associated lymphoid tissues. MZ B-cells can bind to fully glycosylated gp120 and produce specific IgG and IgA, and have a propensity for B regulatory potential, which could help both the fight against HIV and maintenance of low inflammatory conditions reported for HESNs. Here we identify genital MZ-like B-cells, which frequencies are lower in the genital tract of HESNs when compared to HIV-infected CSWs and HIV-uninfected non-CSW women. Furthermore, this coincides with significantly lower genital levels of B lymphocyte stimulator (BLyS/BAFF), known to shape the MZ pool and which overexpression leads to MZ deregulation in HIV-infected progressors. HESN individuals provide an exceptional opportunity to determine important clues for the development of protective devices. Here we show that contained BLyS/BAFF levels are concomitant with natural immunity against HIV, and may prevent dysregulated first-line responses. MZ-like B-cells could be harnessed in preventive strategies viewed at soliciting quick first-line to be adjunct to matured long term protection.
Collapse
Affiliation(s)
- Lyvia Fourcade
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, Canada
| | - Catherine Sabourin-Poirier
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, Canada
| | - Victoire Perraud
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, Canada
| | - Marie-Claude Faucher
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
| | - Josiane Chagnon-Choquet
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
| | - Annie-Claude Labbé
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, Canada
- Département de Microbiologie Médicale et Infectiologie, Hôpital Maisonneuve-Rosemont, Montréal, Canada
| | - Michel Alary
- Centre de recherche du CHU de Québec–Université Laval, Québec, Canada
- Département de Médecine Sociale et Préventive, Université Laval, Québec, Canada
- Institut National de Santé Publique du Québec, Québec, Canada
| | | | - Johanne Poudrier
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, Canada
- * E-mail: (JP); (MR)
| | - Michel Roger
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
- Département de Microbiologie, Infectiologie et Immunologie de l‘Université de Montréal, Montréal, Canada
- * E-mail: (JP); (MR)
| |
Collapse
|
14
|
Bal J, Jung HY, Nguyen LN, Park J, Jang YS, Kim DH. Evaluation of cell-surface displayed synthetic consensus dengue EDIII cells as a potent oral vaccine candidate. Microb Cell Fact 2018; 17:146. [PMID: 30217208 PMCID: PMC6138890 DOI: 10.1186/s12934-018-0994-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/10/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Dengue is a rapidly spreading mosquito borne tropical viral disease affecting hundreds of millions of people across the globe annually. The dengue virus (DENV) includes four genetically distinct serotypes that cause serious life-threatening infections, including dengue hemorrhagic fever/dengue shock syndrome. Dengue vaccine development is complicated by the possibility of vaccine-enhanced severe dengue disease due to antibody-dependent enhancement by pre-existing cross-reactivity, as well as homotypic antibodies. Thus, the development of an efficacious dengue vaccine conferring simultaneous and durable immunity to each of the four DENV serotypes has not yet been developed despite years of research. For mass immunization in deeply affected resource-limited countries, oral vaccination is considered more beneficial than conventional approaches. Therefore, in a continuing effort towards designing economical and potent vaccine candidates, the current study applied yeast surface display technology to develop an oral dengue vaccine candidate using whole recombinant yeast cells displaying the recombinant fusion protein of M cell targeting ligand Co1 fused to the synthetic consensus dengue envelope domain III (scEDIII). Female Balb/c mice were orally fed with recombinant yeast cells and immunogenicity in terms of systemic and mucosal immune responses was monitored. RESULTS Immunofluorescence microscopy with dengue specific antibody and fluorescein isothiocyanate-conjugated anti-mouse IgG antibody clearly showed that recombinant protein Co1-scEDIII-AGA was localized on the cell surface of the respective clones in comparison with scEDIII-Co1 and Mock cells with no fluorescence. Oral dosage applications of surface displayed Co1-scEDIII-AGA stimulated a systemic humoral immune response in the form of dengue-specific serum IgG, as well as a mucosal immune response in the form of secretory immunoglobulin A (sIgA). Antigen-specific B cell responses in isolated lymphoid cells from the spleen and Peyer's patches further supported an elevated mucosal immune response. In addition, surface displayed Co1-scEDIII-AGA feeding elicited strong immune responses in comparison with scEDIII-Co1 and Mock following intraperitoneal booster with purified scEDIII antigen. CONCLUSIONS Surface displayed preparations of Co1-scEDIII-AGA induced strong immunogenicity compared with non-displayed scEDIII-Co1. Prior studies have supported the neutralization potential of scEDIII constructs against all four serotypes. Thus, the oral administration of genetically engineered yeast whole cells displaying biologically active Co1-scEDIII fusion protein without any further processing shows prospective as a potent oral vaccine candidate against dengue viral infection.
Collapse
Affiliation(s)
- Jyotiranjan Bal
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Hee-Young Jung
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Luong Ngoc Nguyen
- Department of Biology, College of Sciences, Hue University, Hue, Vietnam
| | - Jisang Park
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Yong-Suk Jang
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| | - Dae-Hyuk Kim
- Institute for Molecular Biology and Genetics, Department of Molecular Biology, Department of Bioactive Material Sciences, Chonbuk National University, Jeonju, Jeollabuk-do 54896 Republic of Korea
| |
Collapse
|
15
|
Ghosal A. Importance of secreted bacterial RNA in bacterial-host interactions in the gut. Microb Pathog 2017; 104:161-163. [PMID: 28111325 DOI: 10.1016/j.micpath.2017.01.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/12/2017] [Accepted: 01/18/2017] [Indexed: 11/24/2022]
Abstract
Bacteria are ubiquitous in nature and found to be associated with human. Humans are benefited significantly from these associated bacteria. Our understanding is quite limited about these beneficial associations and how bacteria communicate with the host. It is assumed that secreted bacterial products contribute in bacterial-host signaling. A few studies have shown that bacteria secrete RNA into their extracellular medium, and these secreted RNA are capable of altering the host immune response. Multiple studies in eukaryotes have confirmed that secreted RNA can influence the functioning of other cells and their role in the development of several diseases, although not much is known about the composition of secreted bacterial RNA, how they are trafficked and how they impact on the functioning of host cells. By uncovering the beneficial role of secreted bacterial RNA, it would be possible to improve the human health.
Collapse
Affiliation(s)
- Anubrata Ghosal
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, United States.
| |
Collapse
|
16
|
Schoellhammer CM, Schroeder A, Maa R, Lauwers GY, Swiston A, Zervas M, Barman R, DiCiccio AM, Brugge WR, Anderson DG, Blankschtein D, Langer R, Traverso G. Ultrasound-mediated gastrointestinal drug delivery. Sci Transl Med 2016; 7:310ra168. [PMID: 26491078 DOI: 10.1126/scitranslmed.aaa5937] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
There is a significant clinical need for rapid and efficient delivery of drugs directly to the site of diseased tissues for the treatment of gastrointestinal (GI) pathologies, in particular, Crohn's and ulcerative colitis. However, complex therapeutic molecules cannot easily be delivered through the GI tract because of physiologic and structural barriers. We report the use of ultrasound as a modality for enhanced drug delivery to the GI tract, with an emphasis on rectal delivery. Ultrasound increased the absorption of model therapeutics inulin, hydrocortisone, and mesalamine two- to tenfold in ex vivo tissue, depending on location in the GI tract. In pigs, ultrasound induced transient cavitation with negligible heating, leading to an order of magnitude enhancement in the delivery of mesalamine, as well as successful systemic delivery of a macromolecule, insulin, with the expected hypoglycemic response. In a rodent model of chemically induced acute colitis, the addition of ultrasound to a daily mesalamine enema (compared to enema alone) resulted in superior clinical and histological scores of disease activity. In both animal models, ultrasound treatment was well tolerated and resulted in minimal tissue disruption, and in mice, there was no significant effect on histology, fecal score, or tissue inflammatory cytokine levels. The use of ultrasound to enhance GI drug delivery is safe in animals and could augment the efficacy of GI therapies and broaden the scope of agents that could be delivered locally and systemically through the GI tract for chronic conditions such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Carl M Schoellhammer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Avi Schroeder
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Faculty of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Ruby Maa
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gregory Yves Lauwers
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Albert Swiston
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael Zervas
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ross Barman
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Angela M DiCiccio
- The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - William R Brugge
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel Blankschtein
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Giovanni Traverso
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
17
|
Bloemendaal ALA, Buchs NC, George BD, Guy RJ. Intestinal stem cells and intestinal homeostasis in health and in inflammation: A review. Surgery 2016; 159:1237-48. [PMID: 26936524 DOI: 10.1016/j.surg.2016.01.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/06/2016] [Accepted: 01/23/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND The human intestine is a complex group of organs, highly specialized in processing food and providing nutrients to the body. It is under constant threat from microbials and toxins and has therefore developed a number of protective mechanisms. One important mechanism is the constant shedding of epithelial cells into the lumen; another is the production and maintenance of a double-layered mucous boundary in which there is continuous sampling of the luminal microbiota and a persistent presence of antimicrobial enzymes. However, the gut needs commensal bacteria to effectively break down food into absorbable nutrients, which necessitates constant communication between the luminal bacteria and the intestinal immune cells in homeostasis. Disruption of homeostasis, for whatever reason, will give rise to (chronic) inflammation. DISCUSSION Both medical and surgical management of this disruption is discussed.
Collapse
Affiliation(s)
- Alexander L A Bloemendaal
- Colorectal Surgery Department, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom.
| | - Nicolas C Buchs
- Colorectal Surgery Department, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom
| | - Bruce D George
- Colorectal Surgery Department, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom
| | - Richard J Guy
- Colorectal Surgery Department, Oxford University Hospitals, NHS Foundation Trust, Oxford, United Kingdom
| |
Collapse
|
18
|
Hansen JJ. Immune Responses to Intestinal Microbes in Inflammatory Bowel Diseases. Curr Allergy Asthma Rep 2015; 15:61. [PMID: 26306907 DOI: 10.1007/s11882-015-0562-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, are characterized by chronic, T-cell-mediated inflammation of the gastrointestinal tract that can cause significant, lifelong morbidity. Data from both human and animal studies indicate that IBDs are likely caused by dysregulated immune responses to resident intestinal microbes. Certain products from mycobacteria, fungi, and Clostridia stimulate increased effector T cell responses during intestinal inflammation, whereas other bacterial products from Clostridia and Bacteroides promote anti-inflammatory regulatory T cell responses. Antibody responses to bacterial and fungal components may help predict the severity of IBDs. While most currently approved treatments for IBDs generally suppress the patient's immune system, our growing understanding of microbial influences in IBDs will likely lead to the development of new diagnostic tools and therapies that target the intestinal microbiota.
Collapse
Affiliation(s)
- Jonathan J Hansen
- Departments of Medicine, Microbiology and Immunology, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, CB 7032, Chapel Hill, NC, 27599-7032, USA,
| |
Collapse
|
19
|
Berek C. Eosinophils: important players in humoral immunity. Clin Exp Immunol 2015; 183:57-64. [PMID: 26291602 DOI: 10.1111/cei.12695] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2015] [Indexed: 12/13/2022] Open
Abstract
Eosinophils perform numerous tasks. They are involved in inflammatory reactions associated with innate immune defence against parasitic infections and are also involved in pathological processes in response to allergens. Recently, however, it has become clear that eosinophils also play crucial non-inflammatory roles in the generation and maintenance of adaptive immune responses. Eosinophils, being a major source of the plasma cell survival factor APRIL (activation and proliferation-induced ligand), are essential not only for the long-term survival of plasma cells in the bone marrow, but also for the maintenance of these cells in the lamina propria which underlies the gut epithelium. At steady state under non-inflammatory conditions eosinophils are resident cells of the gastrointestinal tract, although only few are present in the major organized lymphoid tissue of the gut - the Peyer's patches (PP). Surprisingly, however, lack of eosinophils abolishes efficient class-switching of B cells to immunoglobulin (Ig)A in the germinal centres of PP. Thus, eosinophils are required to generate and to maintain mucosal IgA plasma cells, and as a consequence their absence leads to a marked reduction of IgA both in serum and in the gut-associated lymphoid tissues (GALT). Eosinophils thus have an essential part in long-term humoral immune protection, as they are crucial for the longevity of antibody-producing plasma cells in the bone marrow and, in addition, for gut immune homeostasis.
Collapse
Affiliation(s)
- C Berek
- B cell Immunology, Deutsches Rheuma Forschungszentrum, Berlin, Germany
| |
Collapse
|
20
|
Alexander KL, Targan SR, Elson CO. Microbiota activation and regulation of innate and adaptive immunity. Immunol Rev 2015; 260:206-20. [PMID: 24942691 DOI: 10.1111/imr.12180] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The human host has coevolved with the collective of bacteria species, termed microbiota, in a complex fashion that affects both innate and adaptive immunity. Differential regulation of regulatory T-cell and effector T-cell responses are a direct result of specific microbial species present within the gut, and this relationship is subject to dysregulation during inflammation and disease. The microbiota varies widely between individuals and has a profound effect on how one reacts to various environmental stimuli, particularly if a person is genetically predisposed to an immune-mediated inflammatory disorder such as inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC). Approximately, half of all CD patients have elevated antibodies to CBir1, a microbiota flagellin common to mice and humans, demonstrating flagellins as immunodominant antigens in the intestines. This review focuses on the use of flagellins as probes to study microbiota-specific responses in the context of health and disease as well as probes of innate and adaptive responses employed by the host to deal with the overwhelming bacterial presence of the microbiota.
Collapse
Affiliation(s)
- Katie L Alexander
- Department of Immunology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | |
Collapse
|
21
|
Chorny A, Cerutti A. Regulation and Function of Mucosal IgA and IgD. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00032-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
22
|
Smith PM, Garrett WS. Gut Microbiota and Intestinal Adaptive Immunity. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00042-2] [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]
|
23
|
McDermott AJ, Huffnagle GB. The microbiome and regulation of mucosal immunity. Immunology 2014; 142:24-31. [PMID: 24329495 DOI: 10.1111/imm.12231] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 11/01/2013] [Accepted: 11/19/2013] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal tract is a mucosal surface constantly exposed to foreign antigens and microbes, and is protected by a vast array of immunologically active structures and cells. Epithelial cells directly participate in immunological surveillance and direction of host responses in the gut and can express numerous pattern recognition receptors, including Toll-like receptor 5 (TLR5), TLR1, TLR2, TLR3, TLR9, and nucleotide oligomerization domain 2, as well as produce chemotactic factors for both myeloid and lymphoid cells following inflammatory stimulation. Within the epithelium and in the underlying lamina propria resides a population of innate lymphoid cells that, following stimulation, can become activated and produce effector cytokines and exert both protective and pathogenic roles during inflammation. Lamina propria dendritic cells play a large role in determining whether the response to a particular antigen will be inflammatory or anti-inflammatory. It is becoming clear that the composition and metabolic activity of the intestinal microbiome, as a whole community, exerts a profound influence on mucosal immune regulation. The microbiome produces short-chain fatty acids, polysaccharide A, α-galactosylceramide and tryptophan metabolites, which can induce interleukin-22, Reg3γ, IgA and interleukin-17 responses. However, much of what is known about microbiome-host immune interactions has come from the study of single bacterial members of the gastrointestinal microbiome and their impact on intestinal mucosal immunity. Additionally, evidence continues to accumulate that alterations of the intestinal microbiome can impact not only gastrointestinal immunity but also immune regulation at distal mucosal sites.
Collapse
Affiliation(s)
- Andrew J McDermott
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | |
Collapse
|
24
|
Cortez VS, Cervantes-Barragan L, Song C, Gilfillan S, McDonald KG, Tussiwand R, Edelson BT, Murakami Y, Murphy KM, Newberry RD, Sibley LD, Colonna M. CRTAM controls residency of gut CD4+CD8+ T cells in the steady state and maintenance of gut CD4+ Th17 during parasitic infection. ACTA ACUST UNITED AC 2014; 211:623-33. [PMID: 24687959 PMCID: PMC3978276 DOI: 10.1084/jem.20130904] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Interactions between cell adhesion molecules CRTAM and Cadm1 regulate the residency and maintenance of CD4+CD8+ and CD4+ T cells in the gut that can influence the immune response to infection. Retention of lymphocytes in the intestinal mucosa requires specialized chemokine receptors and adhesion molecules. We find that both CD4+CD8+ and CD4+ T cells in the intestinal epithelium, as well as CD8+ T cells in the intestinal mucosa and mesenteric lymph nodes, express the cell adhesion molecule class I–restricted T cell–associated molecule (Crtam) upon activation, whereas the ligand of Crtam, cell adhesion molecule 1 (Cadm1), is expressed on gut CD103+DCs. Lack of Crtam–Cadm1 interactions in Crtam−/− and Cadm1−/− mice results in loss of CD4+CD8+ T cells, which arise from mucosal CD4+ T cells that acquire a CD8 lineage expression profile. After acute oral infection with Toxoplasma gondii, both WT and Crtam−/− mice mounted a robust TH1 response, but markedly fewer TH17 cells were present in the intestinal mucosa of Crtam−/− mice. The almost exclusive TH1 response in Crtam−/− mice resulted in more efficient control of intestinal T. gondii infection. Thus, Crtam–Cadm1 interactions have a major impact on the residency and maintenance of CD4+CD8+ T cells in the gut mucosa in the steady state. During pathogenic infection, Crtam–Cadm1 interactions regulate the dynamic equilibrium between newly formed CD4+ T cells and their retention in the gut, thereby shaping representation of disparate CD4+ T cell subsets and the overall quality of the CD4+ T cell response.
Collapse
Affiliation(s)
- Victor S Cortez
- Department of Pathology and Immunology, 2 Department of Internal Medicine, 3 Department of Molecular Microbiology, and 4 Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Monteiro MA, Ma Z, Bertolo L, Jiao Y, Arroyo L, Hodgins D, Mallozzi M, Vedantam G, Sagermann M, Sundsmo J, Chow H. Carbohydrate-based Clostridium difficile vaccines. Expert Rev Vaccines 2013; 12:421-31. [PMID: 23560922 DOI: 10.1586/erv.13.9] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clostridium difficile is responsible for thousands of deaths each year and a vaccine would be welcomed, especially one that would disrupt bacterial maintenance, colonization and persistence in carriers and convalescent patients. Structural explorations at the University of Guelph (ON, Canada) discovered that C. difficile may express three phosphorylated polysaccharides, named PSI, PSII and PSIII; this review captures our recent efforts to create vaccines based on these glycans, especially PSII, the common antigen that has precipitated immediate attention. The authors describe the design and immunogenicity of vaccines composed of raw polysaccharides and conjugates thereof. So far, it has been observed that anti-PSII antibodies can be raised in farm animals, mice and hamster models; humans and horses carry anti-PSII IgA and IgG antibodies from natural exposure to C. difficile, respectively; phosphate is an indispensable immunogenic epitope and vaccine-induced PSII antibodies recognize PSII on C. difficile outer surface.
Collapse
|
26
|
Lopatin U, Blutt SE, Conner ME, Kelsall BL. Lymphotoxin alpha-deficient mice clear persistent rotavirus infection after local generation of mucosal IgA. J Virol 2013; 87:524-30. [PMID: 23097456 PMCID: PMC3536402 DOI: 10.1128/jvi.01801-12] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 10/16/2012] [Indexed: 11/20/2022] Open
Abstract
Rotavirus is a major cause of pediatric diarrheal illness worldwide. To explore the role of organized intestinal lymphoid tissues in infection by and immunity to rotavirus, lymphotoxin alpha-deficient (LTα(-/-)) mice that lack Peyer's patches and mesenteric lymph nodes were orally infected with murine rotavirus. Systemic rotavirus was cleared within 10 days in both LTα(-/-) and wild-type mice, and both strains developed early and sustained serum antirotavirus antibody responses. However, unlike wild-type mice, which resolved the intestinal infection within 10 days, LTα(-/-) mice shed fecal virus for approximately 50 days after inoculation. The resolution of fecal virus shedding occurred concurrently with induction of intestinal rotavirus-specific IgA in both mouse strains. Induction of intestinal rotavirus-specific IgA in LTα(-/-) mice correlated with the (late) appearance of IgA-producing plasma cells in the small intestine. This, together with the absence of rotavirus-specific serum IgA, implies that secretory rotavirus-specific IgA was produced locally. These findings indicate that serum IgG responses are insufficient and imply that local intestinal IgA responses are important for the clearance of rotavirus from intestinal tissues. Furthermore, they show that while LTα-dependent lymphoid tissues are important for the generation of IgA-producing B cells in the intestine, they are not absolutely required in the setting of rotavirus infection. Moreover, the induction of local IgA-producing B cell responses can occur late after infection and in an LTα-independent manner.
Collapse
Affiliation(s)
- Uri Lopatin
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sarah E. Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Margaret E. Conner
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Brian L. Kelsall
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| |
Collapse
|
27
|
Deng Y, Cui H, Peng X, Fang J, Zuo Z, Wang K, Cui W, Wu B. Changes of IgA+ cells and cytokines in the cecal tonsil of broilers fed on diets supplemented with vanadium. Biol Trace Elem Res 2012; 147:149-55. [PMID: 22270623 DOI: 10.1007/s12011-012-9330-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Accepted: 01/10/2012] [Indexed: 11/25/2022]
Abstract
The cecal tonsil of broiler is known as a secondary lymphoid tissue, which is involved in antigen-specific humoral immune responses. The purpose of this study was to investigate the effects of dietary vanadium on the tissue distribution and quantity of immunoglobulin A-positive (IgA(+)) cell in the cecal tonsil by immunohistochemistry. Simultaneously, the changes in interleukin-6 (IL-6), interleukin-10 (IL-10), interferon gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) contents in the cecal tonsil were also quantified by enzyme-linked immunosorbent assay (ELISA). A total of 420 one-day-old avian broilers were divided into six groups and fed on a corn-soybean basal diet (control diet) or the same diet supplemented respectively with 5, 15, 30, 45, and 60 mg/kg of vanadium in the form of ammonium metavanadate for 42 days. The results showed that the population of the IgA(+) cells in the cecal tonsil were significantly lower (p < 0.05 or p < 0.01) in the 45 and 60 mg/kg groups than that in the control group. Meanwhile, IL-10, IFN-γ and TNF-α contents in the cecal tonsil were significantly decreased (p < 0.05 or p < 0.01) in the 30, 45 and 60 mg/kg groups in comparison with those of the control group. However, IL-6 content in the cecal tonsil was only decreased (p < 0.05 or p < 0.01) in 60 mg/kg at 14 and 28 days of age. In conclusion, dietary vanadium in excess of 30 mg/kg reduced the numbers of the IgA(+) cells and changed the contents of the abovementioned cytokines in the cecal tonsil, which may finally impact the function of local mucosal humoral immunity in broilers.
Collapse
Affiliation(s)
- Yuanxin Deng
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Ya'an, China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Spencer J, Klavinskis LS, Fraser LD. The human intestinal IgA response; burning questions. Front Immunol 2012; 3:108. [PMID: 22593756 PMCID: PMC3349913 DOI: 10.3389/fimmu.2012.00108] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 04/17/2012] [Indexed: 12/14/2022] Open
Abstract
The title of this special topic invites us to identify areas in the field of IgA biology that are uncertain or in need of clarification. The inductive phase of the human intestinal IgA response has been a controversial area for some years. Therefore, to structure this review, we have identified key questions that are debated in this field. We have provided explanations of the origins of the uncertainties and have provided our own reasoned answers to the questions we pose.
Collapse
Affiliation(s)
- Jo Spencer
- Peter Gorer Department of Immunobiology, King’s College London School of Medicine at Guy’s King’s College and St. Thomas’ HospitalsLondon, UK
| | - Linda S. Klavinskis
- Peter Gorer Department of Immunobiology, King’s College London School of Medicine at Guy’s King’s College and St. Thomas’ HospitalsLondon, UK
| | - Louise D. Fraser
- Peter Gorer Department of Immunobiology, King’s College London School of Medicine at Guy’s King’s College and St. Thomas’ HospitalsLondon, UK
| |
Collapse
|
29
|
Cerutti A, Cols M, Gentile M, Cassis L, Barra CM, He B, Puga I, Chen K. Regulation of mucosal IgA responses: lessons from primary immunodeficiencies. Ann N Y Acad Sci 2012; 1238:132-44. [PMID: 22129060 DOI: 10.1111/j.1749-6632.2011.06266.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Adaptive co-evolution of mammals and bacteria has led to the establishment of complex commensal communities on mucosal surfaces. In spite of having available a wealth of immune-sensing and effector mechanisms capable of triggering inflammation in response to microbial intrusion, mucosal immune cells establish an intimate dialogue with microbes to generate a state of hyporesponsiveness against commensals and active readiness against pathogens. A key component of this homeostatic balance is IgA, a noninflammatory antibody isotype produced by mucosal B cells through class switching. This process involves activation of B cells by IgA-inducing signals originating from mucosal T cells, dendritic cells, and epithelial cells. Here, we review the mechanisms by which mucosal B cells undergo IgA diversification and production and discuss how the study of primary immunodeficiencies facilitates better understanding of mucosal IgA responses in humans.
Collapse
Affiliation(s)
- Andrea Cerutti
- Municipal Institute for Medical Research-Hospital del Mar, Barcelona, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Svensson-Frej M. Immunobiology of intestinal eosinophils - a dogma in the changing? J Innate Immun 2011; 3:565-76. [PMID: 21860213 DOI: 10.1159/000328799] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 04/19/2011] [Indexed: 12/29/2022] Open
Abstract
Infiltration of eosinophils into the intestinal mucosa is a typical hallmark of antiparasite immune responses and inflammatory disorders of the intestinal tract, and eosinophils are thought to contribute to these processes by release of their cytotoxic granule content. However, utilizing novel tools to study eosinophils, it has been recognized that eosinophils are constitutively present in the gastrointestinal tract. In addition, as the dogmatic antiparasite function of eosinophils has proven difficult to document experimentally, it has become increasingly clear that eosinophils are likely to have a more complex role than previously appreciated. Thus, the prevailing dogma of eosinophils merely as antiparasitic effector cells is changing. Instead, it has been suggested that eosinophils can contribute also to several other processes in the intestinal mucosa, e.g. local tissue homeostasis and adaptive immune responses. This review describes the current knowledge regarding the characteristics and functions of intestinal eosinophils, and the regulation of eosinophil trafficking to the intestinal mucosa during the steady state and inflammation. Finally, potential additional and new roles of intestinal eosinophils in the intestinal mucosal immune system are discussed.
Collapse
|
31
|
Mesin L, Di Niro R, Thompson KM, Lundin KEA, Sollid LM. Long-lived plasma cells from human small intestine biopsies secrete immunoglobulins for many weeks in vitro. THE JOURNAL OF IMMUNOLOGY 2011; 187:2867-74. [PMID: 21841131 DOI: 10.4049/jimmunol.1003181] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To understand the biology of Ab-secreting cells in the human small intestine, we examined Ab production of intestinal biopsies kept in culture. We found sustained IgA and IgM secretion as well as viable IgA- or IgM-secreting cells after >4 wk of culture. The Ab-secreting cells were nonproliferating and expressing CD27 and CD138, thus having a typical plasma cell phenotype. Culturing of biopsies without tissue disruption gave the highest Ab production and plasma cell survival suggesting that the environment regulates plasma cell longevity. Cytokine profiling of the biopsy cultures demonstrated a sustained presence of IL-6 and APRIL. Blocking of the activity of endogenous APRIL and IL-6 with BCMA-Fc and anti-human IL-6 Ab demonstrated that both these factors were essential for plasma cell survival and Ab secretion in the biopsy cultures. This study demonstrates that the human small intestine harbors a population of nonproliferating plasma cells that are instructed by the microenvironment for prolonged survival and Ab secretion.
Collapse
Affiliation(s)
- Luka Mesin
- Center for Immune Regulation, University of Oslo, Q3 N-0027 Oslo, Norway.
| | | | | | | | | |
Collapse
|
32
|
Suzuki K, Kawamoto S, Maruya M, Fagarasan S. GALT: organization and dynamics leading to IgA synthesis. Adv Immunol 2011; 107:153-85. [PMID: 21034974 DOI: 10.1016/b978-0-12-381300-8.00006-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since its discovery more than four decades ago, immunoglobulin (Ig) A has been the subject of continuous and intensive studies. The major concepts derived were that the precursors of IgA plasma cells are generated in follicular organized structures with the help of T cells and the secreted IgAs provide protection against mucosal pathogens. However, only recently we began to appreciate that IgAs play key roles in regulation of bacterial communities in the intestine and that the repertoire of gut microbiota is closely linked to the proper functioning of the immune system. In this review, we highlight the complex and dynamic mutualistic relationships between bacteria and immune cells and discuss the sites and pathways leading to IgA synthesis in gut-associated lymphoid tissues (GALT).
Collapse
Affiliation(s)
- Keiichiro Suzuki
- Research Center for Allergy and Immunology, RIKEN Yokohama Tsurumi, Yokohama, Japan
| | | | | | | |
Collapse
|
33
|
Eosinophils are required for the maintenance of plasma cells in the bone marrow. Nat Immunol 2011; 12:151-9. [PMID: 21217761 DOI: 10.1038/ni.1981] [Citation(s) in RCA: 389] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 12/06/2010] [Indexed: 12/21/2022]
Abstract
Plasma cells are of crucial importance for long-term immune protection. It is thought that long-lived plasma cells survive in specialized niches in the bone marrow. Here we demonstrate that bone marrow eosinophils localized together with plasma cells and were the key providers of plasma cell survival factors. In vitro, eosinophils supported the survival of plasma cells by secreting the proliferation-inducing ligand APRIL and interleukin-6 (IL-6). In eosinophil-deficient mice, plasma cell numbers were much lower in the bone marrow both at steady state and after immunization. Reconstitution experiments showed that eosinophils were crucial for the retention of plasma cells in the bone marrow. Moreover, depletion of eosinophils induced apoptosis in long-lived bone marrow plasma cells. Our findings demonstrate that the long-term maintenance of plasma cells in the bone marrow requires eosinophils.
Collapse
|
34
|
Abstract
Understanding the mechanisms underlying the induction of immunity in the gastrointestinal mucosa following oral immunization and the cross-talk between mucosal and systemic immunity should expedite the development of vaccines to diminish the global burden caused by enteric pathogens. Identifying an immunological correlate of protection in the course of field trials of efficacy, animal models (when available), or human challenge studies is also invaluable. In industrialized country populations, live attenuated vaccines (e.g. polio, typhoid, and rotavirus) mimic natural infection and generate robust protective immune responses. In contrast, a major challenge is to understand and overcome the barriers responsible for the diminished immunogenicity and efficacy of the same enteric vaccines in underprivileged populations in developing countries. Success in developing vaccines against some enteric pathogens has heretofore been elusive (e.g. Shigella). Different types of oral vaccines can selectively or inclusively elicit mucosal secretory immunoglobulin A and serum immunoglobulin G antibodies and a variety of cell-mediated immune responses. Areas of research that require acceleration include interaction between the gut innate immune system and the stimulation of adaptive immunity, development of safe yet effective mucosal adjuvants, better understanding of homing to the mucosa of immunologically relevant cells, and elicitation of mucosal immunologic memory. This review dissects the immune responses elicited in humans by enteric vaccines.
Collapse
Affiliation(s)
- Marcela F Pasetti
- Center for Vaccine Development, University of Maryland School of Medicine, 685 West Baltimore St., Room 480, Baltimore, MD 21201, USA.
| | | | | | | |
Collapse
|
35
|
Abstract
Mucosal surfaces are colonized by large communities of commensal bacteria and represent the primary site of entry for pathogenic agents. To prevent microbial intrusion, mucosal B cells release large amounts of immunoglobulin (Ig) molecules through multiple follicular and extrafollicular pathways. IgA is the most abundant antibody isotype in mucosal secretions and owes its success in frontline immunity to its ability to undergo transcytosis across epithelial cells. In addition to translocating IgA onto the mucosal surface, epithelial cells educate the mucosal immune system as to the composition of the local microbiota and instruct B cells to initiate IgA responses that generate immune protection while preserving immune homeostasis. Here we review recent advances in our understanding of the cellular interactions and signaling pathways governing IgA production at mucosal surfaces and discuss new findings on the regulation and function of mucosal IgD, the most enigmatic isotype of our mucosal antibody repertoire.
Collapse
Affiliation(s)
- Andrea Cerutti
- ICREA, Catalan Institute for Research and Advanced Studies, Barcelona Biomedical Research Park, Spain.
| | | | | |
Collapse
|
36
|
Strugnell RA, Wijburg OLC. The role of secretory antibodies in infection immunity. Nat Rev Microbiol 2010; 8:656-67. [PMID: 20694027 DOI: 10.1038/nrmicro2384] [Citation(s) in RCA: 209] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The mucosal secretory immune system provides an important primary defence against disease, as studies of humans with mucosal humoral immunodeficiencies suggest that the absence of secretory immunoglobulin A leads to an increase in mucosal infections. However, the infection risks posed do not seem to provide the evolutionary drive to retain constitutive secretion of often 'hard won' protein, suggesting that secretory antibodies may have some other important function (or functions). This Review examines the evidence that secretory antibodies provide an important defence against infection in specific animal models and explores complementary explanations for the evolution of the secretory immune system.
Collapse
Affiliation(s)
- Richard A Strugnell
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, Victoria, VIC 3010 Australia.
| | | |
Collapse
|
37
|
Bergqvist P, Stensson A, Lycke NY, Bemark M. T cell-independent IgA class switch recombination is restricted to the GALT and occurs prior to manifest germinal center formation. THE JOURNAL OF IMMUNOLOGY 2010; 184:3545-53. [PMID: 20207993 DOI: 10.4049/jimmunol.0901895] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recently, we reported that CD40(-/-) mice, exhibiting exclusively T cell-independent IgA class switch recombination (CSR), demonstrated near normal levels of IgA plasma cells in the gut lamina propria (LP), despite the complete lack of germinal centers (GCs). In this study, we have extended our analysis focusing on how to reconcile these findings using flow cytometry and molecular markers for IgA CSR. In agreement with our previous results with small intestinal LP, the colon LP was found to host IgA CSR only when lymphoid follicles were present. Thus, no IgA CSR was observed in the nonorganized colon LP. By contrast, the Peyer's patch (PP) was the dominant IgA CSR site in both CD40(-/-) and wild type (WT) mice, and they both hosted similar levels of mRNA expression for B cell activating factor of the TNF family, a proliferation inducing ligand, and inducible NO synthase, potential switch-factors for IgA. Unexpectedly, we found that PP B cells undergoing IgA CSR were GL7-intermediate. These cells had not undergone somatic hypermutations (SHMs), whereas GL7-high cells in WT PP, which exhibited GCs, were heavily mutated. Moreover, IgA plasma cells in the LP of CD40(-/-) mice demonstrated few mutations in their Ig V regions, whereas WT LP B cells from different sites showed extensive SHMs, which were also clonally related. Therefore, IgA CSR can occur in PP at a stage preceding manifest GC (GL7-intermediate), whereas SHM require GC formations (GL7-high). These findings reconcile that IgA CSR can occur in PP in the absence of GC with the fact that CD40(-/-) mice host near normal levels of IgA plasma cells in the LP.
Collapse
Affiliation(s)
- Peter Bergqvist
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | | |
Collapse
|
38
|
Abstract
The past 20 years have seen a growing interest over the control of adaptive immune responses by the innate immune system. In particular, considerable attention has been paid to the mechanisms by which antigen-primed dendritic cells orchestrate the differentiation of T cells. Additional studies have elucidated the pathways followed by T cells to initiate immunoglobulin responses in B cells. In this review, we discuss recent advances on the mechanisms by which intestinal bacteria, epithelial cells, dendritic cells, and macrophages cross talk with intestinal T cells and B cells to induce frontline immunoglobulin A class switching and production.
Collapse
Affiliation(s)
- Alejo Chorny
- Department of Medicine, The Immunology Institute, Mount Sinai School of Medicine, New York, NY, USA
| | | | | |
Collapse
|
39
|
He B, Xu W, Cerutti A. Comment on "Gut-associated lymphoid tissue contains the molecular machinery to support T-cell-dependent and T-cell-independent class switch recombination". Mucosal Immunol 2010; 3:92-4; author reply 94-5. [PMID: 20016479 DOI: 10.1038/mi.2009.125] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
40
|
Nijnik A, Pistolic J, Wyatt A, Tam S, Hancock REW. Human Cathelicidin Peptide LL-37 Modulates the Effects of IFN-γ on APCs. THE JOURNAL OF IMMUNOLOGY 2009; 183:5788-98. [DOI: 10.4049/jimmunol.0901491] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
41
|
Fujihashi K, Kiyono H. Mucosal immunosenescence: new developments and vaccines to control infectious diseases. Trends Immunol 2009; 30:334-43. [DOI: 10.1016/j.it.2009.04.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2009] [Revised: 04/08/2009] [Accepted: 04/08/2009] [Indexed: 11/28/2022]
|
42
|
Abstract
The gut mucosa is exposed to a large community of commensal bacteria that are required for the processing of nutrients and the education of the local immune system. Conversely, the gut immune system generates innate and adaptive responses that shape the composition of the local microbiota. One striking feature of intestinal adaptive immunity is its ability to generate massive amounts of noninflammatory immunoglobulin A (IgA) antibodies through multiple follicular and extrafollicular pathways that operate in the presence or absence of cognate T-B cell interactions. Here we discuss the role of intestinal IgA in host-commensal mutualism, immune protection, and tolerance and summarize recent advances on the role of innate immune cells in intestinal IgA production.
Collapse
Affiliation(s)
- Andrea Cerutti
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, and Weill Graduate School of Medical Sciences of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology (IEO), Via Ripamonti 435, Milan 20141, Italy
| |
Collapse
|
43
|
Abstract
IgA class switching is the process whereby B cells acquire the expression of IgA, the most abundant antibody isotype in mucosal secretions. IgA class switching occurs via both T-cell-dependent and T-cell-independent pathways, and the antibody targets both pathogenic and commensal microorganisms. This Review describes recent advances indicating that innate immune recognition of microbial signatures at the epithelial-cell barrier is central to the selective induction of mucosal IgA class switching. In addition, the mechanisms of IgA class switching at follicular and extrafollicular sites within the mucosal environment are summarized. A better understanding of these mechanisms may help in the development of more effective mucosal vaccines.
Collapse
Affiliation(s)
- Andrea Cerutti
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, and Weill Graduate School of Medical Sciences of Cornell University, 1300 York Avenue, New York, New York 10021, USA.
| |
Collapse
|
44
|
The Society for Mucosal Immunology and Nature Publishing Group bring you Mucosal Immunology. Mucosal Immunol 2008; 1:5-7. [PMID: 19079154 DOI: 10.1038/mi.2007.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|