1
|
Pracht K, Wittner J, Kagerer F, Jäck HM, Schuh W. The intestine: A highly dynamic microenvironment for IgA plasma cells. Front Immunol 2023; 14:1114348. [PMID: 36875083 PMCID: PMC9977823 DOI: 10.3389/fimmu.2023.1114348] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/23/2023] [Indexed: 02/18/2023] Open
Abstract
To achieve longevity, IgA plasma cells require a sophisticated anatomical microenvironment that provides cytokines, cell-cell contacts, and nutrients as well as metabolites. The intestinal epithelium harbors cells with distinct functions and represents an important defense line. Anti-microbial peptide-producing paneth cells, mucus-secreting goblet cells and antigen-transporting microfold (M) cells cooperate to build a protective barrier against pathogens. In addition, intestinal epithelial cells are instrumental in the transcytosis of IgA to the gut lumen, and support plasma cell survival by producing the cytokines APRIL and BAFF. Moreover, nutrients are sensed through specialized receptors such as the aryl hydrocarbon receptor (AhR) by both, intestinal epithelial cells and immune cells. However, the intestinal epithelium is highly dynamic with a high cellular turn-over rate and exposure to changing microbiota and nutritional factors. In this review, we discuss the spatial interplay of the intestinal epithelium with plasma cells and its potential contribution to IgA plasma cell generation, homing, and longevity. Moreover, we describe the impact of nutritional AhR ligands on intestinal epithelial cell-IgA plasma cell interaction. Finally, we introduce spatial transcriptomics as a new technology to address open questions in intestinal IgA plasma cell biology.
Collapse
Affiliation(s)
- Katharina Pracht
- Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jens Wittner
- Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fritz Kagerer
- Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Schuh
- Division of Molecular Immunology, Department of Internal Medicine 3, Nikolaus-Fiebiger-Center, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Immune Impairment Associated with Vitamin A Deficiency: Insights from Clinical Studies and Animal Model Research. Nutrients 2022; 14:nu14235038. [PMID: 36501067 PMCID: PMC9738822 DOI: 10.3390/nu14235038] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/20/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Vitamin A (VA) is critical for many biological processes, including embryonic development, hormone production and function, the maintenance and modulation of immunity, and the homeostasis of epithelium and mucosa. Specifically, VA affects cell integrity, cytokine production, innate immune cell activation, antigen presentation, and lymphocyte trafficking to mucosal surfaces. VA also has been reported to influence the gut microbiota composition and diversity. Consequently, VA deficiency (VAD) results in the imbalanced production of inflammatory and immunomodulatory cytokines, intestinal inflammation, weakened mucosal barrier functions, reduced reactive oxygen species (ROS) and disruption of the gut microbiome. Although VAD is primarily known to cause xerophthalmia, its role in the impairment of anti-infectious defense mechanisms is less defined. Infectious diseases lead to temporary anorexia and lower dietary intake; furthermore, they adversely affect VA status by interfering with VA absorption, utilization and excretion. Thus, there is a tri-directional relationship between VAD, immune response and infections, as VAD affects immune response and predisposes the host to infection, and infection decreases the intestinal absorption of the VA, thereby contributing to secondary VAD development. This has been demonstrated using nutritional and clinical studies, radiotracer studies and knockout animal models. An in-depth understanding of the relationship between VAD, immune response, gut microbiota and infections is critical for optimizing vaccine efficacy and the development of effective immunization programs for countries with high prevalence of VAD. Therefore, in this review, we have comprehensively summarized the existing knowledge regarding VAD impacts on immune responses to infections and post vaccination. We have detailed pathological conditions associated with clinical and subclinical VAD, gut microbiome adaptation to VAD and VAD effects on the immune responses to infection and vaccines.
Collapse
|
3
|
An expanding stage for commensal microbes in host immune regulation. Cell Mol Immunol 2017; 14:339-348. [PMID: 28065939 DOI: 10.1038/cmi.2016.64] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/12/2016] [Accepted: 10/12/2016] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal commensal microbiota is a concentrated mix of microbial life forms, including bacteria, fungi, archaea and viruses. These life forms are targets of host antimicrobial defense in order to establish a homeostatic symbiosis inside the host. However, they are also instrumental in shaping the functions of our immune system via a diverse set of communication mechanisms. In the gut, T helper 17, regulatory T and B cells are continuously tuned by specific microbial strains and metabolic processes. These cells in return help to establish a mutually beneficial exchange with the gut microbial contents. Imbalances in this symbiosis lead to dysregulations in the host's ability to control infections and the development of autoimmune diseases. In addition, the commensal microbiota has a significant and obligatory role in shaping both gut intrinsic and distal lymphoid organs, casting a large impact on the overall immune landscape in the host. This review discusses the major components of the microbial community in the gut and how its members collectively and individually exert regulatory roles in the host immune system and lymphoid structure development, as well as the functions of several major immune cell types.
Collapse
|
4
|
The dual nature of retinoic acid in pemphigus and its therapeutic potential: Special focus on all-trans Retinoic Acid. Int Immunopharmacol 2016; 36:180-186. [PMID: 27156125 DOI: 10.1016/j.intimp.2016.04.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/17/2016] [Accepted: 04/20/2016] [Indexed: 01/13/2023]
Abstract
The efficient treatment of pemphigus with no certain side effect remained a controversial issue. Although there are various options for controlling disease severity, the majority of them may cause serious side effects. Retinoic acid (RA), an active metabolite converted from vitamin A, plays an active role in immune functions. Effects of RA, especially all-trans-Retinoic Acid (ATRA) on different types of cells involved in immune responses were analyzed in vitro and in vivo. RAs could affect the differentiation of T helper (Th) cells, B cells responses, stabilization of both natural regulatory T cells (nTregs) and regulatory B cells (Bregs) populations, and regulating the expression of critical genes in immune responses. The role of RA, based on major immune cells involved in pemphigus has not been addressed so far. In this study, we sought to determine the possible effects of RA, with a special focus on ATRA in pemphigus. All the evidences of ATRA effects on the immune system were collected and their association with the pemphigus was analyzed. According to the previous results, ATRA causes a decline in Th17 populations; increase in CD4+ induced regulatory T cells (iTregs), stabilization of nTregs, and promotion of suppressive B cells, which are critical in the improvement of pemphigus. Nevertheless, it also causes shifting of the Th1:Th2 balance toward Th2 cells, which is not favorable for pemphigus patients. In conclusion, ATRA acts via different ways in pemphigus. Due to increase in the suppressive function via iTregs, nTregs, and Bregs, it is suggested that patients with pemphigus may benefit from systemic ATRA therapy. To clarify this issue, further studies, such as clinical trials are needed.
Collapse
|
5
|
Surman SL, Penkert RR, Jones BG, Sealy RE, Hurwitz JL. Vitamin Supplementation at the Time of Immunization with a Cold-Adapted Influenza Virus Vaccine Corrects Poor Mucosal Antibody Responses in Mice Deficient for Vitamins A and D. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2016; 23:219-27. [PMID: 26740391 PMCID: PMC4783424 DOI: 10.1128/cvi.00739-15] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 12/28/2015] [Indexed: 12/21/2022]
Abstract
Vitamin A and D deficiencies and insufficiencies are prevalent worldwide in developed and developing countries. Vitamin metabolites are functionally intertwined in that they are high-affinity ligands for related receptors of the nuclear receptor superfamily. The effects of vitamin A deficiencies (VAD) on antibody responses to respiratory virus vaccines have already been demonstrated. Of particular concern was the reduction in IgA, a first line of defense against pathogens in the respiratory tract. Here, we describe the individual and combined effects of vitamin A and D deficiencies in mice immunized with an attenuated influenza virus vaccine. Relative to VAD, vitamin D deficiency (VDD) had a limited effect, but double deficiencies for vitamins A and D (VAD+VDD) further reduced antibody responses in the respiratory tract. The administration of supplemental vitamins A and D to VAD+VDD mice at the time of vaccination restored responses in a dose-dependent manner. Results suggest that vitamin supplementation programs may be beneficial in a clinical setting to promote healthy immune responses to respiratory virus vaccines in vitamin-deficient individuals.
Collapse
Affiliation(s)
- S L Surman
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - R R Penkert
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - B G Jones
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - R E Sealy
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - J L Hurwitz
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
6
|
Fan X, Liu S, Liu G, Zhao J, Jiao H, Wang X, Song Z, Lin H. Vitamin A Deficiency Impairs Mucin Expression and Suppresses the Mucosal Immune Function of the Respiratory Tract in Chicks. PLoS One 2015; 10:e0139131. [PMID: 26422233 PMCID: PMC4589363 DOI: 10.1371/journal.pone.0139131] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Accepted: 09/08/2015] [Indexed: 02/03/2023] Open
Abstract
The chicken immune system is immature at the time of hatching. The development of the respiratory immune system after hatching is vital to young chicks. The aim of this study was to investigate the effect of dietary vitamin A supplement levels on respiratory mucin and IgA production in chicks. In this study, 120 one-day-old broiler chicks were randomly divided into 4 groups consisting of three replicates of 10 broilers and subjected to dietary vitamin A supplement levels of 0, 1,500, 6,000, or 12,000 IU/kg for seven days. Compared with control birds, vitamin A supplementation significantly increased the mucin and IgA levels in the bronchoalveolar lavage fluid (BALF) as well as the IgA level in serum. In the lungs, vitamin A supplementation downregulated TNF-α and EGFR mRNA expression. The TGF-β and MUC5AC mRNA expression levels were upregulated by vitamin A supplementation at a dose of 6,000 IU/kg, and the IL-13 mRNA expression level was increased at the 12,000 IU/kg supplement level. Vitamin A deficiency (control) significantly decreased the mRNA expression levels of MUC2, IgA, EGFR, IL-13 and TGF-β in trachea tissue. Histological section analysis revealed that the number of goblet cells in the tracheal epithelium was less in the 0 and 12,000 IU/kg vitamin A supplement groups than in the other groups. In conclusion, vitamin A deficiency suppressed the immunity of the airway by decreasing the IgA and mucin concentrations in neonatal chicks. This study suggested that a suitable level of vitamin A is essential for the secretion of IgA and mucin in the respiratory tract by regulating the gene expression of cytokines and epithelial growth factors.
Collapse
Affiliation(s)
- Xiaoxiao Fan
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, China
| | - Shaoqiong Liu
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, China
| | - Guanhua Liu
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, China
| | - Jingpeng Zhao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, China
| | - Hongchao Jiao
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, China
| | - Xiaojuan Wang
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, China
| | - Zhigang Song
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, China
- * E-mail: (HL); (ZS)
| | - Hai Lin
- Department of Animal Science, Shandong Agricultural University, Shandong Key Lab for Animal Biotechnology and Disease Control, Taian, Shandong, 271018, China
- * E-mail: (HL); (ZS)
| |
Collapse
|
7
|
Kang SH, Jin BR, Kim HJ, Seo GY, Jang YS, Kim SJ, An SJ, Park SR, Kim WS, Kim PH. Lactoferrin Combined with Retinoic Acid Stimulates B1 Cells to Express IgA Isotype and Gut-homing Molecules. Immune Netw 2015; 15:37-43. [PMID: 25713507 PMCID: PMC4338266 DOI: 10.4110/in.2015.15.1.37] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/23/2015] [Accepted: 01/26/2015] [Indexed: 12/03/2022] Open
Abstract
It is well established that TGF-β1 and retinoic acid (RA) cause IgA isotype switching in mice. We recently found that lactoferrin (LF) also has an activity of IgA isotype switching in spleen B cells. The present study explored the effect of LF on the Ig production by mouse peritoneal B cells. LF, like TGF-β1, substantially increased IgA production in peritoneal B1 cells but little in peritoneal B2 cells. In contrast, LF increased IgG2b production in peritoneal B2 cells much more strongly than in peritoneal B1 cells. LF in combination with RA further enhanced the IgA production and, interestingly, this enhancement was restricted to IgA isotype and B1 cells. Similarly, the combination of the two molecules also led to expression of gut homing molecules α4β7 and CCR9 on peritoneal B1 cells, but not on peritoneal B2 cells. Thus, these results indicate that LF and RA can contribute to gut IgA response through stimulating IgA isotype switching and expression of gut-homing molecules in peritoneal B1 cells.
Collapse
Affiliation(s)
- Seong-Ho Kang
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Bo-Ra Jin
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Hyeon-Jin Kim
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Goo-Young Seo
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Young-Saeng Jang
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Sun-Jin Kim
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Sun-Jin An
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| | - Seok-Rae Park
- Department of Microbiology, College of Medicine, Konyang University, Daejeon 302-718, Korea
| | - Woan-Sub Kim
- Department of Animal Life and Environmental Science, College of Agriculture and Life Science, Hankyong National University, Anseong 456-749, Korea
| | - Pyeung-Hyeun Kim
- Department of Molecular Bioscience, School of Biomedical Science, Kangwon National University, Chuncheon 200-701, Korea
| |
Collapse
|
8
|
Retinoic acid acts as a selective human IgA switch factor. Hum Immunol 2014; 75:923-9. [DOI: 10.1016/j.humimm.2014.06.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 03/10/2014] [Accepted: 06/23/2014] [Indexed: 11/22/2022]
|
9
|
Rudraraju R, Jones BG, Surman SL, Sealy RE, Thomas PG, Hurwitz JL. Respiratory tract epithelial cells express retinaldehyde dehydrogenase ALDH1A and enhance IgA production by stimulated B cells in the presence of vitamin A. PLoS One 2014; 9:e86554. [PMID: 24466150 PMCID: PMC3899288 DOI: 10.1371/journal.pone.0086554] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 12/15/2013] [Indexed: 11/18/2022] Open
Abstract
Morbidity and mortality due to viral infections are major health concerns, particularly when individuals are vitamin A deficient. Vitamin A deficiency significantly impairs mucosal IgA, a first line of defense against virus at its point of entry. Previous reports have suggested that CD11c(Hi) dendritic cells (DCs) of the gastrointestinal tract produce retinaldehyde dehydrogenase (ALDH1A), which metabolizes vitamin A precursors to retinoic acid to support normal mucosal immunity. Given that the upper respiratory tract (URT) and gastrointestinal tract share numerous characteristics, we asked if the CD11c(Hi) DCs of the URT might also express ALDH1A. To address this question, we examined both CD11c(Hi) test cells and CD11c(Lo/neg) control cells from nasal tissue. Surprisingly, the CD11c(Lo/neg) cells expressed more ALDH1A mRNA per cell than did the CD11c(Hi) cells. Further evaluation of CD11c(Lo/neg) populations by PCR and staining of respiratory tract sections revealed that epithelial cells were robust producers of both ALDH1A mRNA and protein. Moreover, CD11c(Lo/neg) cells from nasal tissue (and a homogeneous respiratory tract epithelial cell line) enhanced IgA production by lipopolysaccharide (LPS)-stimulated splenocyte cultures in the presence of the retinoic acid precursor retinol. Within co-cultures, there was increased expression of MCP-1, IL-6, and GM-CSF, the latter two of which were necessary for IgA upregulation. All three cytokines/chemokines were expressed by the LPS-stimulated respiratory tract epithelial cell line in the absence of splenocytes. These data demonstrate the autonomous potential of respiratory tract epithelial cells to support vitamin A-mediated IgA production, and encourage the clinical testing of intranasal vitamin A supplements in vitamin A deficient populations to improve mucosal immune responses toward respiratory tract pathogens and vaccines.
Collapse
Affiliation(s)
- Rajeev Rudraraju
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Bart G. Jones
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Sherri L. Surman
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Robert E. Sealy
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Julia L. Hurwitz
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| |
Collapse
|
10
|
Kandasamy S, Chattha KS, Vlasova AN, Saif LJ. Prenatal vitamin A deficiency impairs adaptive immune responses to pentavalent rotavirus vaccine (RotaTeq®) in a neonatal gnotobiotic pig model. Vaccine 2013; 32:816-24. [PMID: 24380684 DOI: 10.1016/j.vaccine.2013.12.039] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 12/06/2013] [Accepted: 12/12/2013] [Indexed: 11/16/2022]
Abstract
Vitamin A deficiency (VAD) is associated with increased childhood mortality and morbidity in impoverished Asian and African countries, but the impact of VAD on rotavirus (RV) vaccine or infection is poorly understood. We assessed effects of gestational and dietary induced pre- and post-natal VAD and vitamin A supplementation on immune responses to a pentavalent rotavirus vaccine, RotaTeq(®) in a neonatal gnotobiotic pig model. Vaccine efficacy was assessed against virulent G1P[8] human rotavirus (HRV) challenge. VAD and vitamin A sufficient (VAS) piglets were derived from dietary VAD and VAS sows, respectively. VAD piglets had significantly lower levels of hepatic vitamin A compared to that of VAS piglets. RotaTeq(®)-vaccinated VAD piglets had 350-fold higher fecal virus shedding titers compared to vaccinated VAS piglets post-challenge. Only 25% of vaccinated non-vitamin A supplemented VAD piglets were protected against diarrhea compared with 100% protection rate in vaccinated non-supplemented VAS piglets post-challenge. Intestinal HRV specific immune responses were compromised in VAD piglets. Vaccinated VAD piglets had significantly lower ileal HRV IgG antibody secreting cell (ASC) responses (pre-challenge) and duodenal HRV IgA ASC responses (post-challenge) compared to vaccinated VAS piglets. Also, intestinal HRV IgA antibody titers were 11-fold lower in vaccinated VAD compared to vaccinated VAS piglets post-challenge. Persistently elevated levels of IL-8, a pro-inflammatory mediator, and lower IL-10 responses (anti-inflammatory) in vaccinated VAD compared to VAS piglets suggest more severe inflammatory responses in VAD piglets post-challenge. Moreover higher IFN-γ responses pre-challenge were observed in VAD compared to VAS piglets. The impaired vaccine-specific intestinal antibody responses and decreased immunoregulatory cytokine responses coincided with reduced protective efficacy of the RV vaccine against virulent HRV challenge in VAD piglets. In conclusion, VAD impaired antibody responses to RotaTeq(®) and vaccine efficacy. Oral supplementation of 100,000 IU vitamin A concurrent with RV vaccine failed to increase the vaccine efficacy in VAD piglets.
Collapse
Affiliation(s)
- Sukumar Kandasamy
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691-4096, USA.
| | - Kuldeep S Chattha
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691-4096, USA.
| | - Anastasia N Vlasova
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691-4096, USA.
| | - Linda J Saif
- Food Animal Health Research Program, Department of Veterinary Preventive Medicine, Ohio Agricultural Research and Development Center, The Ohio State University, 1680 Madison Avenue, Wooster, OH 44691-4096, USA.
| |
Collapse
|
11
|
Seo GY, Jang YS, Kim HA, Lee MR, Park MH, Park SR, Lee JM, Choe J, Kim PH. Retinoic acid, acting as a highly specific IgA isotype switch factor, cooperates with TGF-β1 to enhance the overall IgA response. J Leukoc Biol 2013; 94:325-35. [PMID: 23744644 DOI: 10.1189/jlb.0313128] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The present study demonstrates that RA has activity of an IgA switch factor and is more specific than TGF-β1. RA independently caused only IgA switching, whereas TGF-β1 caused IgA and IgG2b switching. We found that RA increased IgA production and that this was a result of its ability to increase the frequency of IgA-secreting B cell clones. Increased IgA production was accompanied by an increase of GLTα. RA activity was abrogated by an antagonist of the RAR. Additionally, RA affected intestinal IgA production in mice. Surprisingly, RA, in combination with TGF-β1, notably enhanced not only IgA production and GLTα expression but also CCR9 and α4β7 expression on B cells. These results suggest that RA selectively induces IgA isotype switching through RAR and that RA and TGF-β have important effects on the overall gut IgA antibody response.
Collapse
Affiliation(s)
- Goo-Young Seo
- School of Bioscience and Biotechnology, Kangwon National University, Chuncheon 200-701, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Surman SL, Rudraraju R, Sealy R, Jones B, Hurwitz JL. Vitamin A deficiency disrupts vaccine-induced antibody-forming cells and the balance of IgA/IgG isotypes in the upper and lower respiratory tract. Viral Immunol 2012; 25:341-4. [PMID: 22813425 DOI: 10.1089/vim.2012.0023] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vaccination by intranasal (IN) inoculation with a replication-competent virus forms the basis of licensed and novel candidate respiratory viral vaccines (e.g., the cold-adapted influenza virus vaccine). A positive global impact of vaccination depends on vaccine efficacy in developing countries where dietary deficiencies are commonplace. The current study was designed using Sendai virus (SeV) as a model respiratory viral vaccine to test antibody-forming cell (AFC) residence and isotype expression in the context of a vitamin A deficiency (VAD). Samples were taken 1 mo after vaccination when AFCs generally reach their peak in healthy animals. In control animals on a healthy diet, SeV induced an antibody response with a relative bias toward IgA in the upper respiratory tract (URT, as sampled by nasal wash), and IgG in the lower respiratory tract (LRT, as sampled by bronchoalveolar lavage [BAL]). In the context of VAD, the SeV-specific IgA antibodies in the nasal wash were significantly reduced in favor of enhanced IgG antibodies in the BAL. When AFCs were examined in diffuse nasal-associated lymphoid tissues (d-NALT), lungs, cervical lymph nodes (CLN), and mediastinal lymph nodes (MLN), a similar pattern emerged. AFCs were most frequent in the d-NALT and most expressed IgA in control mice. In the context of VAD, these IgA-producing AFCs were significantly reduced in number, skewing the natural balance of IgA and IgG. Taken together, the results show that the VAD diet, which is well known for its association with immune defects in the gut, significantly alters AFC induction and isotype expression in the respiratory tract.
Collapse
Affiliation(s)
- Sherri L Surman
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | |
Collapse
|
13
|
Cassani B, Villablanca EJ, De Calisto J, Wang S, Mora JR. Vitamin A and immune regulation: role of retinoic acid in gut-associated dendritic cell education, immune protection and tolerance. Mol Aspects Med 2011; 33:63-76. [PMID: 22120429 DOI: 10.1016/j.mam.2011.11.001] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 11/01/2011] [Accepted: 11/10/2011] [Indexed: 01/27/2023]
Abstract
The vitamin A (VA) metabolite all-trans retinoic acid (RA) plays a key role in mucosal immune responses. RA is produced by gut-associated dendritic cells (DC) and is required for generating gut-tropic lymphocytes and IgA-antibody-secreting cells (IgA-ASC). Moreover, RA modulates Foxp3(+) regulatory T cell (T(REG)) and Th17 effector T cell differentiation. Thus, although RA could be used as an effective "mucosal adjuvant" in vaccines, it also appears to be required for establishing intestinal immune tolerance. Here we discuss the roles proposed for RA in shaping intestinal immune responses and tolerance at the gut mucosal interface. We also focus on recent data exploring the mechanisms by which gut-associated DC acquire RA-producing capacity.
Collapse
Affiliation(s)
- Barbara Cassani
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | |
Collapse
|
14
|
Matzinger P, Kamala T. Tissue-based class control: the other side of tolerance. Nat Rev Immunol 2011; 11:221-30. [PMID: 21350581 DOI: 10.1038/nri2940] [Citation(s) in RCA: 261] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In this Essay, we offer a new perspective on how immune responses are regulated. We do not cover how they are turned on and off, but focus instead on the second major aspect of an immune response: the control of effector class. Although it is generally thought that the class of an immune response is tailored to fit the invading pathogen, we suggest here that it is primarily tailored to fit the tissue in which the response occurs. To this end, we cover such topics as the nature of T helper (T(H)) cell subsets (current and yet to be discovered), the nature of privileged sites, the difference between oral tolerance and oral vaccination, why the route of immunization matters, whether the T(H)1-type response is really the immune system's primary defense, and whether there might be a different role for some regulatory T cells.
Collapse
Affiliation(s)
- Polly Matzinger
- Laboratory of Cellular and Molecular Immunology, T-Cell Tolerance and Memory Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
15
|
Abstract
Vitamin A (also called retinol), absorbed in the intestine and stored mainly in the liver and fat, is normally maintained at significant concentrations in the human blood plasma. Vitamin A is constitutively metabolized at high levels in certain tissues such as the small intestine and eyes. Retinoic acid (RA) produced at high levels in the intestine plays important roles in mucosal immunity and immune tolerance. RA at basal levels is required for immune cell survival and activation. During immune responses, enzymes metabolizing vitamin A are induced in certain types of immune cells such as dendritic cells (DCs) and tissue cells for induced production of RA. As a result, induced gradients of RA are formed during immune responses in the body. RA regulates gene expression, differentiation, and function of diverse immune cells. The cells under the influence of RA in terms of differentiation include myeloid cells such as neutrophils, macrophages, and DCs. Also included are lymphoid cells such as effector T cells, regulatory T cells, and B cells. Our current understanding of the function of RA in regulation of these immune cells is reviewed in this chapter.
Collapse
|
16
|
Mucida D, Park Y, Cheroutre H. From the diet to the nucleus: vitamin A and TGF-beta join efforts at the mucosal interface of the intestine. Semin Immunol 2009; 21:14-21. [PMID: 18809338 PMCID: PMC2643336 DOI: 10.1016/j.smim.2008.08.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Revised: 08/03/2008] [Accepted: 08/05/2008] [Indexed: 10/21/2022]
Abstract
The vitamin A metabolites, including retinoic acid (RA), form ligands for retinoic acid-related nuclear receptors and together they play pleiotropic roles in various biological processes. Recently, we described that RA also functions as a key modulator of transforming growth factor-beta (TGF-beta)-driven immune deviation, capable of suppressing the differentiation of interleukin-17 secreting T helper cells (T(H)17) and conversely promoting the generation of Foxp3(+) T regulatory (Treg) cells. This review will focus on the role of RA in the reciprocal TGF-beta-driven differentiation of T(H)17 and Treg and on the importance of such regulatory mechanism to control a functional immune system, in particular at the mucosal interface of the intestine.
Collapse
Affiliation(s)
- Daniel Mucida
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
17
|
Mora JR, von Andrian UH. Role of retinoic acid in the imprinting of gut-homing IgA-secreting cells. Semin Immunol 2008; 21:28-35. [PMID: 18804386 DOI: 10.1016/j.smim.2008.08.002] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 08/07/2008] [Accepted: 08/09/2008] [Indexed: 12/20/2022]
Abstract
Antibody-secreting cells (ASCs) lodging in the mucosa of the small intestine are derived from activated B cells that are thought to arise in gut-associated lymphoid tissues (GALT). Upon leaving the GALT, B cells return to the blood where they must express the gut-homing receptors alpha4beta7 and CCR9 in order to emigrate into the small bowel. Recent evidence indicates that gut-associated dendritic cells (DCs) in GALT induce gut-homing receptors on B cells via a mechanism that depends on the vitamin A metabolite retinoic acid (RA). In addition, although ASC associated with other mucosal tissues secrete IgA in an RA-independent fashion, the presence of high levels of RA in intestine and GALT can promote B cell class switching to IgA and thus, boost the production of IgA in the intestinal mucosa. Here, we discuss the role of RA in the imprinting of gut-homing ASC and the evidence linking RA with the generation of intestinal IgA-ASCs.
Collapse
Affiliation(s)
- J Rodrigo Mora
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, GRJ-815, Boston, MA 02114, USA.
| | | |
Collapse
|
18
|
Boehm MF, Heyman RA, Patel S, Stein RB, Nagpal S. Section Review: Retinoids: Biological Function and Use in the Treatment of Dermatological Diseases: Pulmonary-Allergy, Dermatological, Gastrointestinal & Arthritis. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.4.7.593] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
19
|
Abstract
Most antibody-secreting cells (ASCs) in mucosal tissues produce immunoglobulin A (IgA), the most abundant immunoglobulin in the body and the main class of antibody found in secretions. IgA-ASCs differentiate in the mucosal-associated lymphoid tissues and are usually considered as a homogeneous population of cells. However, IgA-ASCs that travel to the small intestine have unique characteristics in terms of their migratory requirements. These IgA-ASCs require the homing molecules alpha4beta7 and CCR9 to interact with their ligands, mucosal addressin cell adhesion molecule-1 and CCL25, which are constitutively expressed in the small intestine. Indeed, recent work has shown that IgA-ASCs specific for the small bowel are generated under different conditions as compared with IgA-ASCs in other mucosal compartments. Moreover, the mechanisms inducing IgA class switching may also vary according to the tissue where IgA-ASCs differentiate. Here we describe the mechanisms involved in the differentiation of IgA-ASCs in mucosal compartments, in particular those involved in the generation of gut-homing IgA-ASCs.
Collapse
|
20
|
Mora JR. Homing imprinting and immunomodulation in the gut: role of dendritic cells and retinoids. Inflamm Bowel Dis 2008; 14:275-89. [PMID: 17924560 DOI: 10.1002/ibd.20280] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lymphocyte migration is at the heart of chronic inflammatory ailments, including inflammatory bowel disease (IBD). Whereas naïve lymphocytes migrate to all secondary lymphoid organs, they are mostly excluded from nonlymphoid peripheral tissues. Upon activation, lymphocytes change their pattern of adhesion receptors and acquire the capacity to migrate to extralymphoid tissues. Antigen-experienced T cells are subdivided into different subsets based on their expression of homing receptors that favor their accumulation in specific tissues, such as the skin and the gut mucosa. B cells and antibody-secreting cells (ASC) also show tissue-tropism, which is somewhat correlated with the class of immunoglobulin that they produce. In fact, IgA-ASC are located in mucosal tissues, where they produce IgA, the main class of antibodies found in secretions. Although IgA-ASC are usually considered as a homogeneous pool of cells, those located in the small bowel have some unique migratory characteristics, suggesting that they are generated under different conditions as compared to IgA-ASC in other mucosal compartments. Foxp3(+) regulatory T cells (T(REG)) can also exhibit tissue-specific migratory potential and recent evidence suggests that T(REG) can be imprinted with gut-specific homing. Moreover, foxp3(+) T(REG) are enriched in the small bowel lamina propria, where they can be generated locally. The present review addresses our current understanding of how tissue-specific homing is acquired and modulated on T cells, B cells, and ASC, with a special emphasis on the intestinal mucosa. Harnessing these mechanisms could offer novel, effective, and more specific therapeutic strategies in IBD.
Collapse
Affiliation(s)
- J Rodrigo Mora
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
21
|
Mora JR, Iwata M, Eksteen B, Song SY, Junt T, Senman B, Otipoby KL, Yokota A, Takeuchi H, Ricciardi-Castagnoli P, Rajewsky K, Adams DH, von Andrian UH. Generation of Gut-Homing IgA-Secreting B Cells by Intestinal Dendritic Cells. Science 2006; 314:1157-60. [PMID: 17110582 DOI: 10.1126/science.1132742] [Citation(s) in RCA: 774] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Normal intestinal mucosa contains abundant immunoglobulin A (IgA)-secreting cells, which are generated from B cells in gut-associated lymphoid tissues (GALT). We show that dendritic cells (DC) from GALT induce T cell-independent expression of IgA and gut-homing receptors on B cells. GALT-DC-derived retinoic acid (RA) alone conferred gut tropism but could not promote IgA secretion. However, RA potently synergized with GALT-DC-derived interleukin-6 (IL-6) or IL-5 to induce IgA secretion. Consequently, mice deficient in the RA precursor vitamin A lacked IgA-secreting cells in the small intestine. Thus, GALT-DC shape mucosal immunity by modulating B cell migration and effector activity through synergistically acting mediators.
Collapse
Affiliation(s)
- J Rodrigo Mora
- CBR Institute for Biomedical Research and Department of Pathology, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Tu Y, Jerry DJ, Pazik B, Smith Schneider S. Sensitivity to DNA damage is a common component of hormone-based strategies for protection of the mammary gland. Mol Cancer Res 2005; 3:435-42. [PMID: 16123139 DOI: 10.1158/1541-7786.mcr-05-0038] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An early full-term pregnancy significantly reduces the risk of getting breast cancer in women. In animals, this protection can be mimicked by a short-term exposure to physiologic doses of estrogen plus progesterone. Sensitization of p53 and up-regulation of transforming growth factor beta are believed to be important aspects of the mechanism by which protection is imparted. Little is known, however, about the use of this pathway in response to other chemopreventive agents. In this article, we investigated the ability of retinoids, such as 9-cis retinoic acid, all-trans retinoic acid, and N-4-hydroxyphenylretinamide (4-HPR), to sensitize the ductal epithelial cells of virgin mammary glands to DNA damage responses. Using a whole-organ culture system, we observed enhanced cell death in response to gamma-irradiation in the virgin tissues treated with retinoids for 72 hours. These retinoids were partially dependent on p53 and transforming growth factor beta to exert their radiosensitizing effects. However, 4-HPR seemed to sensitize other cells or activate these pathways in a different manner as costimulation with ovarian hormones and 4-HPR was additive, whereas coculture of ovarian hormones and the natural retinoids did not increase amount of death. Taken together, these data suggest that sensitization of the mammary epithelium to p53-dependent apoptosis is a common pathway, which is engaged by retinoids as well as ovarian hormones.
Collapse
Affiliation(s)
- Yifan Tu
- Molecular and Cellular Biology Program, University of Massachussetts at Amherst, Amherst, MA 01107, USA.
| | | | | | | |
Collapse
|
23
|
Sato Y, Akiyama H, Suganuma H, Watanabe T, Nagaoka MH, Inakuma T, Goda Y, Maitani T. The Feeding of .BETA.-Carotene Down-Regulates Serum IgE Levels and Inhibits the Type I Allergic Response in Mice. Biol Pharm Bull 2004; 27:978-84. [PMID: 15256726 DOI: 10.1248/bpb.27.978] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Feed containing beta-carotene was administered orally to BALB/c mice immunized intraperitoneally with ovalbumin (OVA) for approximately 1 month. The titers of OVA-specific IgE, OVA-specific IgG1 and OVA-specific IgG2a in the mouse sera were determined. The OVA-specific IgE titer and OVA-specific IgG1 titer by mice fed beta-carotene were significantly inhibited. On the other hand, the OVA-specific IgG2a titer in mice fed beta-carotene was significantly greater than those of control mice. The OVA-specific IgE suppression of beta-carotene feeding was dose-dependent. We also examined the effect of fed beta-carotene on active systemic anaphylaxis. Feeding beta-carotene to mice immunized with OVA inhibited the immediate reduction of the body temperature induced by antigen stimulation. Furthermore, the increase in serum histamine in the mice fed beta-carotene under active systemic anaphylaxis was lower than in controls. We then examined the pattern of cytokine production by spleen cells from mice followed by restimulation with OVA in vitro. The spleen cells from the mice fed beta-carotene produced more IFN-gamma, IL-12 and IL-2 than those from the control group. In contrast, the spleen cells from the mice fed beta-carotene produced less IL-4, IL-5, IL-6, IL-10 than those from the control group. Furthermore, analysis of IFN-gamma mRNA levels of the splenocytes using the real-time quantitative RT-PCR technique revealed higher levels in the splenocytes from the mice fed beta-carotene. These findings suggest that feeding beta-carotene improves the helper T cell (T(H))1-T(H)2 balance, inhibiting specific IgE and IgG1 production and antigen-induced anaphylactic response.
Collapse
Affiliation(s)
- Yuji Sato
- National Institute of Health Sciences, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Aukrust P, Müller F, Ueland T, Svardal AM, Berge RK, Frøland SS. Decreased vitamin A levels in common variable immunodeficiency: vitamin A supplementation in vivo enhances immunoglobulin production and downregulates inflammatory responses. Eur J Clin Invest 2000; 30:252-9. [PMID: 10692003 DOI: 10.1046/j.1365-2362.2000.00619.x] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Vitamin A has a broad range of immunological effects, and vitamin A deficiency is associated with recurrent infections. Common variable immunodeficiency (CVI) is a group of B-cell deficiency syndromes with impaired antibody production and recurrent bacterial infections as the major manifestations, but the immunological dysfunctions may also include T cells and macrophages. In the present study we examined the possible role of vitamin A deficiency in CVI. PATIENTS AND METHODS We analysed plasma vitamin A levels in 20 CVI patients and 16 controls, and examined the relationships between vitamin A and clinical, immunological and metabolic parameters in CVI. In the six CVI patients with the lowest vitamin A levels we also studied the effect of vitamin A supplementation in vivo on several immunological functions in these patients. RESULTS (i) The majority of CVI patients had decreased vitamin A levels compared with healthy controls, as found in both cross-sectional and longitudinal testing. (ii) Low vitamin A levels were associated with the occurrence of chronic bacterial infections and splenomegaly as well as high neopterin levels. Decreased levels of carrier protein and malabsorption were not observed. (iii) Vitamin A supplementation in patients with low vitamin A levels resulted in increased interleukin-10 (IL-10) and decreased tumour necrosis factor-alpha (TNFalpha) levels, as found in both plasma and monocyte supernatants, possibly favouring anti-inflammatory net effects. (iv) Vitamin A supplementation in vivo also enhanced anti-CD40-stimulated IgG production, serum IgA levels and phytohaemagglutinin (PHA)-stimulated peripheral blood mononuclear cell (PBMC) proliferation. CONCLUSION A considerable subgroup of CVI patients appears to be characterized by low vitamin A levels. Our findings support a possible role for vitamin A supplementation in CVI, perhaps resulting in enhanced immunoglobulin synthesis and downregulated inflammatory responses.
Collapse
Affiliation(s)
- P Aukrust
- University of Oslo, Oslo; University of Bergen, Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
25
|
Tokuyama H, Tokuyama Y. The regulatory effects of all-trans-retinoic acid on isotype switching: retinoic acid induces IgA switch rearrangement in cooperation with IL-5 and inhibits IgG1 switching. Cell Immunol 1999; 192:41-7. [PMID: 10066345 DOI: 10.1006/cimm.1998.1438] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
All-trans-retinoic acid (RA) can induce germline Calpha transcription in LPS-stimulated murine mu(+)B-cells by a TGF-beta-independent mechanism. In the present study, we examined whether RA can further drive the IgA switching process to Smu-Salpha switch rearrangement by DC-PCR. RA alone could not induce switch rearrangement but required the cooperation of IL-5. RA has another effect on isotype switching; RA strongly inhibits IL-4-dependent IgG1 and IgE production. To analyze the mechanism of IgG1 inhibition, we tested whether RA can inhibit IL-4-dependent Smu-Sgamma1 switch rearrangement. IL-4 by itself could induce Smu-Sgamma1 switch rearrangement in LPS-stimulated mu(+)B-cells. Addition of RA inhibited this reaction. RA also showed an inhibitory effect on the preceding step, i.e., Igamma1Cgamma1 transcription. Therefore, RA inhibition of Smu-Sgamma1 switch rearrangement was regulated at the level of germline Cgamma1 transcription. We further analyzed the amounts of both Igamma1Cgamma1 and IalphaCalpha expressed in LPS-stimulated B-cells exposed to mixtures of the two switch inducers, RA and IL-4, at various concentrations and found that the two transcripts were regulated antagonistically. These results indicated that RA can regulate isotype switching at the level of germline transcription and directs switching to IgA with the help of IL-5 and inhibits IgG1 switching.
Collapse
Affiliation(s)
- H Tokuyama
- Department of Molecular Immunology, Cancer Research Institute, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa-ken, 920-0934, Japan
| | | |
Collapse
|
26
|
Tokuyama H, Tokuyama Y. Retinoic acid induces the expression of germ-line C alpha transcript mainly by a TGF-beta-independent mechanism. Cell Immunol 1997; 176:14-21. [PMID: 9070313 DOI: 10.1006/cimm.1996.1069] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The effects of retinoic acid (RA) on expression of germ-line transcripts, I alpha C alpha and I gamma 1C gamma 1, and of IgA and IgG1 mRNAs by murine surface IgM-positive B-cells were examined by reverse transcriptase-polymerase chain reaction (RT-PCR). LPS-stimulated B-cells were cultured for 2-3 days in the presence of IL-4 and IL-5 with or without RA. Total RNA was extracted from the cells, and RT-PCR specific for the germ-line transcripts was carried out. RA strongly induced mature germ-line C alpha transcript (I alpha C alpha) at concentrations between 10 and 100 nM. On the other hand, RA completely inhibited IL-4-induced I gamma 1C gamma 1 expression. Significant induction of I alpha C alpha was observed even at a low RA concentration (0.2 nM) in the presence of LPS (1.5-5 micrograms/ml) and without cytokines, and three- to fourfold stimulation of I alpha C alpha induction was seen at 5 nM. I alpha C alpha expression induced by RA (10 nM) and LPS (1.5 micrograms/ml) was not significantly affected by addition of anti-TGF-beta 1 and anti-TGF-beta 2 neutralizing antibodies, although that induced by TGF-beta 1 or TGF-beta 2 was completely inhibited by these antibodies. These results suggest that the major induction pathway of I alpha C alpha was not mediated by active TGF-beta and that RA at physiological concentrations may be involved in IgA isotype switching in vivo in a TGF-beta-independent manner.
Collapse
Affiliation(s)
- H Tokuyama
- Department of Molecular Immunology, Kanazawa University, Japan
| | | |
Collapse
|
27
|
Abstract
A newly synthesized retinoid, Am-80 (4-[5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthalenyl] carbamoxyl) benzoic acid, induced limb reduction defects in rats in vivo. The present study was designed to investigate the mechanisms of its teratogenic action and also to examine whether transforming growth factor-beta (TGF-beta) is involved in limb reduction defects by using a rat limb bud cell culture system. Am-80 inhibited DNA synthesis and chondrogenesis of limb bud cells from day 12 embryos at concentrations of 1-4 ng/ml in a concentration-dependent manner. Quantities of TGF-beta 1 and 2 in culture medium from limb bud cells treated with Am-80 were evaluated using sandwich ELISAs. Although TGF-beta 1 was not detected in conditioned medium with or without Am-80, TGF-beta 2 increased in a concentration-dependent manner at drug concentrations that inhibited DNA synthesis and chondrogenesis. In addition, when TGF-beta 2 was exogenously added to limb bud cell culture medium, the synthesis of glycosaminoglycan (GAGs) was reduced even without Am-80 treatment. Neutralization of TGF-beta 2 with antibody prevented the inhibition of GAGs synthesis induced by Am-80. These results suggest that TGF-betas, especially beta 2, are involved in the inhibition of chondrogenesis caused by Am-80 in limb bud cells.
Collapse
Affiliation(s)
- H Tsuiki
- Developmental Research Laboratories, Shionogi & Co., Ltd., Osaka, Japan
| | | | | |
Collapse
|
28
|
Kuwabara K, Shudo K, Hori Y. Novel synthetic retinoic acid inhibits rat collagen arthritis and differentially affects serum immunoglobulin subclass levels. FEBS Lett 1996; 378:153-6. [PMID: 8549823 DOI: 10.1016/0014-5793(95)01440-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Retinoids affect many biological processes such as cell proliferation, differentiation and morphogenesis, but their effects on arthritic patients and animal models of arthritis are controversial. We tested the effect of a novel synthetic retinoic acid, Am-80 (4-[(5,6,7,8-tetrahydro-5,5,8,8,-tetramethyl-2-naphthalenyl) carbamoyl] benzoic acid), on type-II collagen (CII)-induced arthritis (CIA) in rats. Am-80 markedly suppressed the incidence of arthritis, hindpaw swelling and bone destruction. In contrast, 13-cis-retinoic acid (13-cis-RA) hardly inhibited these CIA symptoms. Moreover, Am-80, but not 13-cis-RA, strongly reduced the serum level of anti-CII antibody and differentially affected the levels of immunoglobulin (Ig) subclasses in vivo: IgG1 and IgG2a levels were decreased, while IgA level was increased without any change in the IgM level. These findings indicate that Am-80 may be one of the lead retinoic acids of a new class of anti-inflammatory agents.
Collapse
Affiliation(s)
- K Kuwabara
- Discovery Research Laboratories II, Shionogi and Co., Osaka, Japan
| | | | | |
Collapse
|
29
|
Zorn NE, Sauro MD. Retinoic acid induces translocation of protein kinase C (PKC) and activation of nuclear PKC (nPKC) in rat splenocytes. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 1995; 17:303-11. [PMID: 7672881 DOI: 10.1016/0192-0561(95)00036-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Retinoic acid (RA), a vitamin A metabolite, has marked effects on growth of normal and malignant cells; however, the exact mechanism of action remains unclear. The effect of two RA analogs, 13-cis-RA and all-trans-RA, on transmembrane signalling processes was investigated in rat splenocytes. Treatment of rat splenic cells with these retinoic acid analogs resulted in translocation of protein kinase C (PKC) from the cytosol to the membrane. Previous studies have described nuclear RA receptors (RARs and RXRs) for several species and the biologic activity of RA has been shown to be mediated by specific interaction with these nuclear receptors. Thus, activation of nuclear pool(s) of protein kinase C (nPKC) by RA analogs was also studied. Rat splenocyte nuclei pure by enzymatic and electron microscope criteria demonstrated a biphasic pattern of bell-shaped curves for both cis- and trans-RA with maximum statistically significant peak of phosphate incorporation into endogenous substrates at 10(-16) M cis-RA and 10(-16)-10(-17) M trans-RA. A monoclonal antibody to PKC and the PKC inhibitors, H-7, sphingosine, and staurosporine, blocked the RA-stimulated nuclear phosphorylation. The ability of RA to activate cell membrane PKC resulting in an increase in particulate PKC activity correlates well with the activation of nPKC since the particulate fraction would include nuclear enzyme systems. This ability of RA to activate nPKC and possibly affect the growth status of a cell may provide a missing link to our understanding of the cellular sites of action for this vitamin.
Collapse
Affiliation(s)
- N E Zorn
- Department of Pediatrics, University of South Florida College of Medicine, Tampa 33612, USA
| | | |
Collapse
|
30
|
Tokuyama Y, Tokuyama H. Retinoic acid and steroid hormones regulate IgA production by LPS-stimulated murine spleen cells. IMMUNOPHARMACOLOGY 1994; 28:145-51. [PMID: 8002288 DOI: 10.1016/0162-3109(94)90030-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We examined the regulatory effects of steroid hormones (beta-estradiol and glucocorticoids) on in vitro IgA production. Addition of retinoic acid (RA, 100-500 nM) to the LPS-stimulated spleen cell culture enhanced IgA production (8-22-fold). Simultaneous addition of beta-estradiol, but not testosterone, enhanced the effect of RA synergistically (a further 2-4-fold). In contrast, glucocorticoids inhibited the reaction. The concentration inhibiting IgA production by 50% was 1 nM, 6 nM and 10 nM for dexamethasone, prednisolone and hydrocortisone, respectively. None of the hormones tested alone affected IgA production by LPS-stimulated spleen cells. Hydrocortisone enhanced the IgG1 production by LPS-stimulated spleen cells. This effect was completely abolished by simultaneous addition of RA. These findings indicate that RA can direct the class-switching to IgA in LPS-stimulated spleen cells, and that beta-estradiol and glucocorticoids have positive and negative regulatory effects, respectively, on the IgA production.
Collapse
Affiliation(s)
- Y Tokuyama
- Department of Nuclear Medicine, Faculty of Medicine, Kanazawa University, Ishikawa-ken, Japan
| | | |
Collapse
|