1
|
Liu W, Jiang H, Liu X, Zheng Y, Liu Y, Pan F, Yu F, Li Z, Gu M, Du Q, Li X, Zhang H, Han D. Altered intestinal microbiota enhances adenoid hypertrophy by disrupting the immune balance. Front Immunol 2023; 14:1277351. [PMID: 38090578 PMCID: PMC10715246 DOI: 10.3389/fimmu.2023.1277351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Adenoid hypertrophy (AH) is a common upper respiratory disorder in children. Disturbances of gut microbiota have been implicated in AH. However, the interplay of alteration of gut microbiome and enlarged adenoids remains elusive. Methods 119 AH children and 100 healthy controls were recruited, and microbiome profiling of fecal samples in participants was performed using 16S rRNA gene sequencing. Fecal microbiome transplantation (FMT) was conducted to verify the effects of gut microbiota on immune response in mice. Results In AH individuals, only a slight decrease of diversity in bacterial community was found, while significant changes of microbial composition were observed between these two groups. Compared with HCs, decreased abundances of Akkermansia, Oscillospiraceae and Eubacterium coprostanoligenes genera and increased abundances of Bacteroides, Faecalibacterium, Ruminococcus gnavus genera were revealed in AH patients. The abundance of Bacteroides remained stable with age in AH children. Notably, a microbial marker panel of 8 OTUs were identified, which discriminated AH from HC individuals with an area under the curve (AUC) of 0.9851 in the discovery set, and verified in the geographically different validation set, achieving an AUC of 0.9782. Furthermore, transfer of mice with fecal microbiota from AH patients dramatically reduced the proportion of Treg subsets within peripheral blood and nasal-associated lymphoid tissue (NALT) and promoted the expansion of Th2 cells in NALT. Conclusion These findings highlight the effect of the altered gut microbiota in the AH pathogenesis.
Collapse
Affiliation(s)
- Wenxin Liu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huier Jiang
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiling Liu
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Ministry of Justice, Shanghai, China
| | - Yue Zheng
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanan Liu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fen Pan
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fangyuan Yu
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Li
- Department of Pathology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meizhen Gu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingqing Du
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyan Li
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhang
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dingding Han
- Department of Clinical Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Institute of Pediatric Infection, Immunity, and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Medical School, Guangxi University, Nanning, China
| |
Collapse
|
2
|
Gondhowiardjo SA, Adham M, Rachmadi L, Atmakusuma TD, Tobing DL, Auzan M, Hariyanto AD, Sulaeman D, Permata TBM, Handoko. Immune cells markers within local tumor microenvironment are associated with EBV oncoprotein in nasopharyngeal cancer. BMC Cancer 2022; 22:887. [PMID: 35963999 PMCID: PMC9375267 DOI: 10.1186/s12885-022-09948-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction EBV infection in nasopharyngeal cancer ensued in latent infection mode. In this latent infection various EBV oncoproteins such as EBNA1 and LMP1 was expressed. EBV oncoproteins could theoretically recruit immune cells, which might help to control cancer. Therefore, this study was aimed to elucidate the association with EBV oncoproteins (EBNA1 and LMP1), immune markers (CD4, CD8, and FOXP3) from nasopharyngeal cancer microenvironment with tumor progression. Method Nasopharyngeal biopsy was obtained from patients suspected to have nasopharyngeal cancer. Those samples with microscopically confirmed nasopharyngeal cancer were tested for EBNA1, LMP1, CD4, CD8, and FOXP3 concentration with ELISA, then verified with IHC. Each patient tumor volume was assessed for primary nasopharyngeal tumor volume (GTVp) and neck nodal metastases tumor volume (GTVn). Correlation test with Spearman correlation and scatterplot were carried out. Result Total 23 samples with nasopharyngeal cancer were analyzed. There was moderate correlation (ρ = 0.45; p value = 0.032) between LMP1 and GTVp. There was strong correlation (ρ = 0.81; p value < 0.001) between CD8 and GTVp. There was also moderate correlation (ρ = 0.6; p value = 0.002) between FOXP3 and GTVp. The CD8 concentration has moderate correlation with both EBNA1 (ρ = 0.46; p value = 0.026) and LMP1 (ρ = 0.47; p value = 0.023). While FOXP3 has moderate correlation with only LMP1 (ρ = 0.58; p value = 0.004). No correlation was found between all the markers tested here with GTVn. Discussion We found larger primary nasopharyngeal tumor was associated with higher CD8 marker. This was thought due to the presence of abundance CD8 T cells in the nasopharynx, but those abundance CD8 T cells were suspected to be dysfunctional. The nasopharyngeal cancer was also known to upregulate chemokines that could recruit T regulatory FOXP3 cells. Furthermore, T regulatory FOXP3 cells differentiation was induced through several pathways which was triggered by EBNA1. The correlation found in this study could guide further study to understand nasopharyngeal carcinogenesis and the relationship with our immune system.
Collapse
Affiliation(s)
- Soehartati A Gondhowiardjo
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Marlinda Adham
- Department of ENT, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Lisnawati Rachmadi
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Tubagus Djumhana Atmakusuma
- Department of Medical Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Demak Lumban Tobing
- Department of Clinical Pathology, Dharmais National Cancer Hospital, Jakarta - Indonesia, Jl. Letjen S. Parman No. 84-86, Jakarta, Indonesia, 11420
| | - Mahesa Auzan
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Agustinus Darmadi Hariyanto
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Dede Sulaeman
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Tiara Bunga Mayang Permata
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Handoko
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430.
| |
Collapse
|
3
|
Ogawa K, Asano K, Yotsumoto S, Yamane T, Arita M, Hayashi Y, Harada H, Makino-Okamura C, Fukuyama H, Kondo K, Yamasoba T, Tanaka M. Frontline Science: Conversion of neutrophils into atypical Ly6G + SiglecF + immune cells with neurosupportive potential in olfactory neuroepithelium. J Leukoc Biol 2021; 109:481-496. [PMID: 32725843 DOI: 10.1002/jlb.1hi0620-190rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022] Open
Abstract
Neutrophils are generally considered as short-lived, homogenous, and terminally differentiated phagocytes that play crucial roles in conquering infection, although they occasionally cause severe collateral tissue damage or chronic inflammation. Recent reports have indicated that neutrophils also play a protective role in inflammation resolution and tissue repair. However, how terminally differentiated neutrophils have diverse functions remains unclear. Here, we show that neutrophils undergo conversion into Ly6G+ SiglecF+ double-positive cells expressing neurosupportive genes in the olfactory neuroepithelium (OE) under an inflammatory state. Through comprehensive flow cytometric analysis of murine nose, we identified Ly6G+ SiglecF+ double-positive cells that reside only in the OE under steady-state conditions. Double-positive cells were neutrophil-derived cells and increased by more than 10-fold during inflammation or tissue injury. We found that neutrophils infiltrate into the nose to express proinflammatory genes in the acute phase of inflammatory state, and they gradually change their surface markers and gene expression, expressing some neurogenesis-related genes in addition to inflammation related genes in the later phase. As the OE is known to have exceptionally high regeneration capacity as a nervous system, these findings suggest that neutrophils have the potential to contribute neurogenesis after conversion in peripheral nervous tissues, providing a challenge on a classic view of neutrophils as terminally differentiated leukocytes.
Collapse
Affiliation(s)
- Kei Ogawa
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Department of Otolaryngology - Head and Neck Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Kenichi Asano
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Satoshi Yotsumoto
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Tsuyoshi Yamane
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Division of Gastroenterology & Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Arita
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Yoshihiro Hayashi
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hironori Harada
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Chieko Makino-Okamura
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Hidehiro Fukuyama
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kenji Kondo
- Department of Otolaryngology - Head and Neck Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Tatsuya Yamasoba
- Department of Otolaryngology - Head and Neck Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Masato Tanaka
- Laboratory of Immune Regulation, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
4
|
Yamamoto S, Matsuo K, Sakai S, Mishima I, Hara Y, Oiso N, Kawada A, Yoshie O, Nakayama T. P2X receptor agonist enhances tumor-specific CTL responses through CD70+ DC-mediated Th17 induction. Int Immunol 2020; 33:49-55. [PMID: 33027512 DOI: 10.1093/intimm/dxaa068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022] Open
Abstract
Extracellular ATP is known to promote Th17 cell differentiation in the intestinal lamina propria by stimulating CD70+CD11clow dendritic cells (DCs) via P2X receptors (P2XRs). Recent studies have also shown that Th17 cells enhance antitumor immunity by directly promoting proliferation of cytotoxic T lymphocytes (CTLs). These finding led us to test a P2XR agonist, αβ-methylene ATP (αβ-ATP), as a mucosal vaccine adjuvant to promote CTL responses through Th17 induction. We demonstrated that (i) CD70+CD11clow DCs were present in the nasal lamina propria and expressed P2X1R, P2X2R and P2X4R; (ii) CD70+CD11clow DCs isolated from the nasal lamina propria enhanced Th17 cell differentiation of cocultured splenic CD4+ T cells upon stimulation with αβ-ATP; (iii) mice intranasally immunized with ovalbumin (OVA) and αβ-ATP had increased OVA-specific Th17 cells and CTLs in the nasal lamina propria and regional lymph nodes; (iv) mice intranasally immunized with OVA and αβ-ATP also had elevated resistance to E.G7-OVA tumor growth compared with those intranasally immunized with OVA alone; (v) suramin, a broad-range inhibitor of P2 receptors, suppressed the increases of OVA-specific Th17 cells and CTLs in mice intranasally immunized with OVA and αβ-ATP; and (vi) suramin also abrogated the enhanced antitumor immunity of mice intranasally immunized with OVA and αβ-ATP against E.G7-OVA. Collectively, αβ-ATP may be a promising mucosal adjuvant that promotes antigen-specific CTL responses via CD70+CD11clow DC-mediated Th17 induction.
Collapse
Affiliation(s)
- Shinya Yamamoto
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-osaka, Osaka, Japan
| | - Kazuhiko Matsuo
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-osaka, Osaka, Japan
| | - Sho Sakai
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-osaka, Osaka, Japan
| | - Itsuki Mishima
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-osaka, Osaka, Japan
| | - Yuta Hara
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-osaka, Osaka, Japan
| | - Naoki Oiso
- Department of Dermatology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Akira Kawada
- Department of Dermatology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Osamu Yoshie
- Emeritus professor, Kindai University, OsakaSayama, Osaka, Japan.,Health and Kampo Institute, Sendai, Miyagi, Japan
| | - Takashi Nakayama
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Higashi-osaka, Osaka, Japan
| |
Collapse
|
5
|
Matsui M, Nagakubo D, Satooka H, Hirata T. A novel Siglec-F + neutrophil subset in the mouse nasal mucosa exhibits an activated phenotype and is increased in an allergic rhinitis model. Biochem Biophys Res Commun 2020; 526:599-606. [PMID: 32247614 DOI: 10.1016/j.bbrc.2020.03.122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/20/2020] [Indexed: 11/17/2022]
Abstract
Neutrophils are important phagocytic cells for host defense against pathogens. They are rapidly recruited to the site of infection, release antimicrobial peptides and cytokines, and engulf and kill microbes. Neutrophils also accumulate in allergic inflammatory sites. Here we characterized neutrophil accumulation in the nasal mucosa using a mouse model of allergic rhinitis, in which mice were sensitized by intraperitoneal injection of ovalbumin (OVA) and then challenged by intranasal administration of OVA or PBS. In the nasal mucosa of both PBS- and OVA-challenged mice, we found a cell subset expressing the eosinophil marker Siglec-F in the Ly-6G+ neutrophil population. Morphological analysis of the sorted Ly-6G+Siglec-F+ cells revealed that they were devoid of eosinophilic granules in the cytosol and were apparently neutrophils, but compared to conventional Ly-6G+Siglec-F- neutrophils, they had a more lobulated, "botryoid" nucleus. Siglec-F+ neutrophils were barely found in the nasopharynx-associated lymphoid tissue, cervical lymph nodes, the spleen, or blood. Both Siglec-F+ neutrophils and conventional neutrophils showed increased numbers in the nasal mucosa of OVA-challenged mice. Compared to conventional Siglec-F- neutrophils, Siglec-F+ neutrophils exhibited an activated phenotype and enhanced effector functions. Taken together, our findings identify Siglec-F+ neutrophils as a novel neutrophil subset with an activated phenotype that resides specifically in the nasal mucosa.
Collapse
Affiliation(s)
- Makoto Matsui
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| | - Daisuke Nagakubo
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| | - Hiroki Satooka
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| | - Takako Hirata
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| |
Collapse
|
6
|
Generation of protective pneumococcal-specific nasal resident memory CD4 + T cells via parenteral immunization. Mucosal Immunol 2020; 13:172-182. [PMID: 31659300 PMCID: PMC6917870 DOI: 10.1038/s41385-019-0218-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 09/23/2019] [Accepted: 10/04/2019] [Indexed: 02/08/2023]
Abstract
The generation of tissue-resident memory T cells (TRM) is an essential aspect of immunity at mucosal surfaces, and it has been suggested that preferential generation of TRM is one of the principal advantages of mucosally administered vaccines. We have previously shown that antigen-specific, IL-17-producing CD4+ T cells can provide capsular antibody-independent protection against nasal carriage of Streptococcus pneumoniae; but whether pneumococcus-responsive TRM are localized within the nasal mucosa and are sufficient for protection from carriage has not been determined. Here, we show that intranasal administration of live or killed pneumococci to mice generates pneumococcus-responsive IL-17A-producing CD4+ mucosal TRM. Furthermore, we show that these cells are sufficient to mediate long-lived, neutrophil-dependent protection against subsequent pneumococcal nasal challenge. Unexpectedly, and in contrast with the prevailing paradigm, we found that parenteral administration of killed pneumococci also generates protective IL-17A+CD4+ TRM in the nasal mucosa. These results demonstrate a critical and sufficient role of TRM in prevention of pneumococcal colonization, and further that these cells can be generated by parenteral immunization. Our findings therefore have important implications regarding the generation of immune protection at mucosal surfaces by vaccination.
Collapse
|
7
|
Yu YY, Kong W, Yin YX, Dong F, Huang ZY, Yin GM, Dong S, Salinas I, Zhang YA, Xu Z. Mucosal immunoglobulins protect the olfactory organ of teleost fish against parasitic infection. PLoS Pathog 2018; 14:e1007251. [PMID: 30395648 PMCID: PMC6237424 DOI: 10.1371/journal.ppat.1007251] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/15/2018] [Accepted: 10/08/2018] [Indexed: 01/12/2023] Open
Abstract
The olfactory organ of vertebrates receives chemical cues present in the air or water and, at the same time, they are exposed to invading pathogens. Nasal-associated lymphoid tissue (NALT), which serves as a mucosal inductive site for humoral immune responses against antigen stimulation in mammals, is present also in teleosts. IgT in teleosts is responsible for similar functions to those carried out by IgA in mammals. Moreover, teleost NALT is known to contain B-cells and teleost nasal mucus contains immunoglobulins (Igs). Yet, whether nasal B cells and Igs respond to infection remains unknown. We hypothesized that water-borne parasites can invade the nasal cavity of fish and elicit local specific immune responses. To address this hypothesis, we developed a model of bath infection with the Ichthyophthirius multifiliis (Ich) parasite in rainbow trout, Oncorhynchus mykiss, an ancient bony fish, and investigated the nasal adaptive immune response against this parasite. Critically, we found that Ich parasites in water could reach the nasal cavity and successfully invade the nasal mucosa. Moreover, strong parasite-specific IgT responses were detected in the nasal mucus, and the accumulation of IgT+ B-cells was noted in the nasal epidermis after Ich infection. Strikingly, local IgT+ B-cell proliferation and parasite-specific IgT generation were found in the trout olfactory organ, providing new evidence that nasal-specific immune responses were induced locally by a parasitic challenge. Overall, our findings suggest that nasal mucosal adaptive immune responses are similar to those reported in other fish mucosal sites and that an antibody system with a dedicated mucosal Ig performs evolutionary conserved functions across vertebrate mucosal surfaces. The olfactory organ is a vitally important chemosensory organ in vertebrates but it is also continuously stimulated by pathogenic microorganisms in the external environment. In mammals and birds, nasopharynx-associated lymphoid tissue (NALT) is considered one of the first lines of immune defense against inhaled antigens and in bony fish, protecting against water-borne infections. However, although B-cells and immunoglobulins (Igs) have been found in teleost NALT, the defensive mechanisms of parasite-specific immune responses after pathogen challenge in the olfactory organ of teleost fish remain poorly understood. Considering that the NALT of all vertebrates has been subjected to similar evolutionary forces, we hypothesize that mucosal Igs play a critical role in the defense of olfactory systems against parasites. To confirm this hypothesis, we show the local proliferation of IgT+ B-cells and production of pathogen-specific IgT within the nasal mucosa upon parasite infection, indicating that parasite-specific IgT is the main Ig isotype specialized for nasal-adaptive immune responses. From an evolutionary perspective, our findings contribute to expanding our view of nasal immune systems and determining the fate of the host–pathogen interaction.
Collapse
Affiliation(s)
- Yong-Yao Yu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Weiguang Kong
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ya-Xing Yin
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Fen Dong
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Zhen-Yu Huang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Guang-Mei Yin
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Shuai Dong
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Irene Salinas
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Yong-An Zhang
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Zhen Xu
- Department of Aquatic Animal Medicine, College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei, China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- * E-mail:
| |
Collapse
|
8
|
Carrasco-Yepez MM, Campos-Rodríguez R, Reséndiz-Albor AA, Peña-Juárez C, Contis-Montes de Oca A, Arciniega-Martínez IM, Bonilla-Lemus P, Rojas-Hernandez S. Naegleria fowleri immunization modifies lymphocytes and APC of nasal mucosa. Parasite Immunol 2018; 40. [PMID: 29243267 DOI: 10.1111/pim.12508] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/01/2017] [Indexed: 11/28/2022]
Abstract
We investigated whether intranasal immunization with amoebic lysates plus cholera toxin modified the populations of T and B lymphocytes, macrophages and dendritic cells by flow cytometry from nose-associated lymphoid tissue (NALT), cervical lymph nodes (CN), nasal passages (NP) and spleen (SP). In all immunized groups, the percentage of CD4 was higher than CD8 cells. CD45 was increased in B cells from mice immunized. We observed IgA antibody-forming cell (IgA-AFC) response, mainly in NALT and NP. Macrophages from NP and CN expressed the highest levels of CD80 and CD86 in N. fowleri lysates with either CT or CT alone immunized mice, whereas dendritic cells expressed high levels of CD80 and CD86 in all compartment from immunized mice. These were lower than those expressed by macrophages. Only in SP from CT-immunized mice, these costimulatory molecules were increased. These results suggest that N. fowleri and CT antigens are taking by APCs, and therefore, protective immunity depends on interactions between APCs and T cells from NP and CN. Consequently, CD4 cells stimulate the differentiation from B lymphocytes to AFC IgA-positive; antibody that we previously found interacting with trophozoites in the nasal lumen avoiding the N. fowleri attachment to nasal epithelium.
Collapse
Affiliation(s)
- M M Carrasco-Yepez
- Proyecto CyMA, Los Reyes Iztacala, UIICSE, UNAM FES Iztacala, Tlalnepantla, Mexico
| | - R Campos-Rodríguez
- Departamento de Bioquímica, Escuela Superior de Medicina, Laboratorio de Bioquímica, Instituto Politécnico Nacional, Mexico City, Mexico
| | - A A Reséndiz-Albor
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| | - C Peña-Juárez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| | - A Contis-Montes de Oca
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| | - I M Arciniega-Martínez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| | - P Bonilla-Lemus
- Proyecto CyMA, Los Reyes Iztacala, UIICSE, UNAM FES Iztacala, Tlalnepantla, Mexico
| | - S Rojas-Hernandez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Laboratorio de Inmunobiología Molecular y Celular, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
9
|
Bankvall M, Jontell M, Wold A, Östman S. Tissue-specific Differences in Immune Cell Subsets Located in the Naso-oropharyngeal-associated Lymphoid Tissues. Scand J Immunol 2017; 87:15-27. [PMID: 29077981 DOI: 10.1111/sji.12625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 10/21/2017] [Indexed: 01/09/2023]
Abstract
Defining the immune cells within the naso-oropharyngeal-associated lymphoid tissues would promote the development of efficient orally and nasally delivered immunotherapies. The aim was to compare murine antigen-presenting cells (APCs) and T cell subsets in the nose-associated lymphoid tissues (NALT), cervical lymph nodes (CLN), mesenteric lymph nodes (MLN) and peripheral lymph nodes (PLN) using flow cytometry and in vitro proliferation assays. Overall, the NALT contained a higher proportion of APCs and a lower proportion of T cells compared to the CLN, MLN and PLN. The APCs of the NALT more often belonged to the CD11c+ CD11b+ and the CD11cneg CD11b+ subsets as compared to the other sites. Both of these APC populations showed little sign of activation, that is low expression of the markers CD40, CD86 and IAd. Instead, the APCs of the NALT more often co-expressed CX3CR1 and CD206, markers associated with a tolerogenic function. No increase in the proportion of regulatory T cells was observed in the NALT. Instead, the T cells frequently exhibited a memory/effector phenotype, expressing the homing markers α4β7, CCR4 and CCR9, but rarely the naïve phenotype cell surface marker CD45RB. In contrast, the T cells at the other sites were mostly of the naïve phenotype. In addition, cells from the NALT did not proliferate upon in vitro stimulation with Con A, whereas the cells from the other sites did. Taken together, these results suggest that the NALT is primarily an effector site rather than one for activation and differentiation, despite it being regarded as a site of induction.
Collapse
Affiliation(s)
- M Bankvall
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - M Jontell
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - A Wold
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S Östman
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
10
|
Blevins LK, Parsonage D, Oliver MB, Domzalski E, Swords WE, Alexander-Miller MA. A Novel Function for the Streptococcus pneumoniae Aminopeptidase N: Inhibition of T Cell Effector Function through Regulation of TCR Signaling. Front Immunol 2017; 8:1610. [PMID: 29230212 PMCID: PMC5711787 DOI: 10.3389/fimmu.2017.01610] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 11/07/2017] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pneumoniae (Spn) causes a variety of disease states including fatal bacterial pneumonia. Our previous finding that introduction of Spn into an animal with ongoing influenza virus infection resulted in a CD8+ T cell population with reduced effector function gave rise to the possibility of direct regulation by pneumococcal components. Here, we show that treatment of effector T cells with lysate derived from Spn resulted in inhibition of IFNγ and tumor necrosis factor α production as well as of cytolytic granule release. Spn aminopeptidase N (PepN) was identified as the inhibitory bacterial component and surprisingly, this property was independent of the peptidase activity found in this family of proteins. Inhibitory activity was associated with reduced activation of ZAP-70, ERK1/2, c-Jun N-terminal kinase, and p38, demonstrating the ability of PepN to negatively regulate TCR signaling at multiple points in the cascade. These results reveal a novel immune regulatory function for a bacterial aminopeptidase.
Collapse
Affiliation(s)
- Lance K Blevins
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Derek Parsonage
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Melissa B Oliver
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Elizabeth Domzalski
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - W Edward Swords
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Martha A Alexander-Miller
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
11
|
Bankvall M, Östberg AK, Jontell M, Wold A, Östman S. The engagement of oral-associated lymphoid tissues during oral versus gastric antigen administration. Immunology 2017; 149:98-110. [PMID: 27288650 DOI: 10.1111/imm.12633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 05/31/2016] [Accepted: 06/03/2016] [Indexed: 12/12/2022] Open
Abstract
The role of oral-associated lymphoid tissues during induction of oral tolerance still remains elusive. Therefore, the aim was to compare T-cell activation and induction of tolerance to ovalbumin (OVA) presented through either of two routes; deposited into the oral cavity, or the stomach, thereby bypassing the oral cavity. OVA was administered by the oral or gastric route to BALB/c mice that had received OVA-specific DO11.10+ CD4(+) T cells, stained with CellTrace(™) Violet dye, through intravenous injection. Proliferating OVA-specific T cells were detected in the nose-associated lymphoid tissues (NALT) and the cervical, mesenteric and peripheral lymph nodes at different time-points following OVA exposure. OVA-specific T-cell proliferation was initially observed in the NALT 1 hr after oral, but not gastric, administration. However, at day 1, proliferation at this site was also detected after gastric administration and profound proliferation was observed at all sites by day 4. For the oral route the degree of proliferation observed was lower in the peripheral lymph nodes by day 4 compared with the other sites. These results demonstrate a similar activation pattern achieved by the two routes. However, the NALT distinguishes itself as a site of rapid T-cell activation towards fed antigens irrespective of feeding regimen. To evaluate induction of tolerance a semi-effective OVA dose was used, to detect differences in the degree of tolerance achieved. This was performed in a model of OVA-induced airway hypersensitivity. No differences in tolerance induction were observed between the two administration routes.
Collapse
Affiliation(s)
- Maria Bankvall
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Anna-Karin Östberg
- Department of Oral Microbiology and Immunology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Mats Jontell
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Agnes Wold
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Sofia Östman
- Department of Infectious Diseases, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| |
Collapse
|
12
|
Cytokine profile of NALT during acute stress and its possible effect on IgA secretion. Immunol Lett 2017; 188:68-78. [PMID: 28472640 DOI: 10.1016/j.imlet.2017.04.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/27/2017] [Accepted: 04/28/2017] [Indexed: 12/20/2022]
Abstract
Stress stimuli affect the immune system responses that occur at mucosal membranes, particularly IgA secretion. It has been suggested that acute stress increases the levels of IgA and that sympathetic innervation plays an important role in this process. We herein explore in a murine model how acute stress affects the Th1/Th2/Treg cytokine balance in NALT, and the possible role of glucocorticoids in this effect. Nine-week-old male CD1 mice were divided into three groups: unstressed (control), stressed (subjected to 4h of immobilization), and stressed after pretreatment with a single dose of the corticosterone receptor antagonist RU-486. The parameters evaluated included plasma corticosterone and epinephrine, IgA levels in nasal fluid (by ELISA), the percentage of CD19+B220+IgA+ lymphocytes and CD138+IgA+ plasma cells, and the mRNA expression of heavy α chain, J chain and pIgR. Moreover, the gene and protein expression of Th1 cytokines (TNFα, IL-2 and INF-γ), Th2 cytokines (IL-4 and IL-5) and Treg cytokines (IL-10 and TGFβ) were determined in nasal mucosa. The results show that acute stress generated a shift towards the dominance of an anti-inflammatory immune response (Th2 and Treg cytokines), evidenced by a significant rise in the amount of T cells that produce IL4, IL-5 and IL-10. This immune environment may favor IgA biosynthesis by CD138+IgA+ plasma cells, a process mediated mostly by glucocorticoids.
Collapse
|
13
|
Cry1Ac toxin induces macrophage activation via ERK1/2, JNK and p38 mitogen-activated protein kinases. Int J Biochem Cell Biol 2016; 78:106-115. [PMID: 27394658 DOI: 10.1016/j.biocel.2016.06.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 06/15/2016] [Accepted: 06/29/2016] [Indexed: 12/14/2022]
Abstract
The Cry1Ac toxin from Bacillus thuringiensis is used commercially as a bio-insecticide and is expressed in transgenic plants that are used for human and animal consumption. Although it was originally considered innocuous for mammals, the Cry1Ac toxin is not inert and has the ability to induce mucosal and systemic immunogenicity. Herein, we examined whether the Cry1Ac toxin promotes macrophage activation and explored the signalling pathways that may mediate this effect. Treatment of primary and RAW264.7 macrophages with the Cry1Ac toxin resulted in upregulation of the costimulatory molecules CD80, CD86 and ICOS-L and enhanced production of nitric oxide, the chemokine MCP-1 and the proinflammatory cytokines TNF-α and IL-6. Remarkably, the Cry1Ac toxin induced phosphorylation of the mitogen-activated protein kinases (MAPKs) ERK1/2, JNK and p38 and promoted nuclear translocation of nuclear factor-kappa B (NF-κB) p50 and p65. p38 and ERK1/2 MAPKs were involved in this effect, as indicated by the Cry1Ac-induced upregulation of CD80 and IL-6 and TNF-α abrogation by the p38 MAPK inhibitor SB203580. Furthermore, treatment the MEK1/2 kinase inhibitor PD98059 blocked increases in MCP-1 secretion and augmented Cry1Ac-induced ICOS-L upregulation. These data demonstrate the capacity of the Cry1Ac toxin to induce macrophage activation via the MAPK and NF-κB pathways.
Collapse
|
14
|
Upregulated CCL28 expression in the nasal mucosa in experimental allergic rhinitis: Implication for CD4(+) memory T cell recruitment. Cell Immunol 2016; 302:58-62. [PMID: 26868716 DOI: 10.1016/j.cellimm.2016.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 02/01/2016] [Indexed: 11/22/2022]
Abstract
During nasal immune responses, lymphocytes activated in the nasopharynx-associated lymphoid tissue (NALT) are thought to traffic to the nasal mucosa. Here we found a prominent infiltration of CD4(+) memory T cells into the nasal mucosa in a mouse model of allergic rhinitis. CCR3 and CCR10 mRNA was increased in the NALT, and CCR3- or CCR10-expressing CD4(+) T cells were present in the nasal mucosa. CCL28, a chemokine ligand for CCR3 and CCR10, was upregulated in nasal epithelial cells. Our results suggest that memory CD4(+) T cells traffic to the nasal mucosa in a process that may involve CCL28 and its receptors CCR3 and CCR10.
Collapse
|
15
|
Identification and Analysis of Natural Killer Cells in Murine Nasal Passages. PLoS One 2015; 10:e0142920. [PMID: 26575399 PMCID: PMC4648567 DOI: 10.1371/journal.pone.0142920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 10/28/2015] [Indexed: 01/02/2023] Open
Abstract
Background Natural killer (NK) cells in the upper respiratory airways are not well characterized. In the current study, we sought to characterize and functionally assess murine nasal NK cells. Methods Using immunohistochemistry and flow cytometry, we compared the nasal NK cells of Ncr1GFP/+ knock-in mice, whose NK cells produced green fluorescent protein, with their splenic and pulmonary counterparts. In addition, we functionally analyzed the nasal NK cells of these mice in vitro. To assess the in vivo functions of nasal NK cells, C57BL/6 mice depleted of NK cells after treatment with PK136 antibody were nasally infected with influenza virus PR8. Results Immunohistochemical analysis confirmed the presence of NK cells in the lamina propria of nasal mucosa, and flow cytometry showed that these cells were of NK cell lineage. The expression patterns of Ly49 receptor, CD11b/CD27, CD62L and CD69 revealed that nasal NK cells had an immature and activated phenotype compared with that of their splenic and pulmonary counterparts. Effector functions including degranulation and IFN(interferon)-γ production after in vitro stimulation with phorbol 12-myristate-13-acetate plus ionomycin or IL(interleukin)-12 plus IL-18 were dampened in nasal NK cells, and the depletion of NK cells led to an increased influenza virus titer in nasal passages. Conclusions The NK cells of the murine nasal passage belong to the conventional NK cell linage and characteristically demonstrate an immature and activated phenotype. Despite their hyporesponsiveness in vitro, nasal NK cells play important roles in the host defense against nasal influenza virus infection.
Collapse
|
16
|
Sepahi A, Salinas I. The evolution of nasal immune systems in vertebrates. Mol Immunol 2015; 69:131-8. [PMID: 26391349 DOI: 10.1016/j.molimm.2015.09.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/05/2015] [Accepted: 09/06/2015] [Indexed: 11/30/2022]
Abstract
The olfactory organs of vertebrates are not only extraordinary chemosensory organs but also a powerful defense system against infection. Nasopharynx-associated lymphoid tissue (NALT) has been traditionally considered as the first line of defense against inhaled antigens in birds and mammals. Novel work in early vertebrates such as teleost fish has expanded our view of nasal immune systems, now recognized to fight both water-borne and air-borne pathogens reaching the olfactory epithelium. Like other mucosa-associated lymphoid tissues (MALT), NALT of birds and mammals is composed of organized lymphoid tissue (O-NALT) (i.e., tonsils) as well as a diffuse network of immune cells, known as diffuse NALT (D-NALT). In teleosts, only D-NALT is present and shares most of the canonical features of other teleost MALT. This review focuses on the evolution of NALT in vertebrates with an emphasis on the most recent findings in teleosts and lungfish. Whereas teleost are currently the most ancient group where NALT has been found, lungfish appear to be the earliest group to have evolved primitive O-NALT structures.
Collapse
Affiliation(s)
- Ali Sepahi
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Irene Salinas
- Center for Evolutionary and Theoretical Immunology, Department of Biology, University of New Mexico, Albuquerque, NM, USA.
| |
Collapse
|
17
|
Cholesteryl Pullulan Encapsulated TNF-α Nanoparticles Are an Effective Mucosal Vaccine Adjuvant against Influenza Virus. BIOMED RESEARCH INTERNATIONAL 2015; 2015:471468. [PMID: 26421290 PMCID: PMC4569761 DOI: 10.1155/2015/471468] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 01/05/2023]
Abstract
We encapsulated tumor necrosis factor-α (TNF-α), a major proinflammatory cytokine, into cholesteryl pullulan (CHP) to prepare TNF/CHP nanoparticles. In this report, we describe the immune-enhancing capability of the nanoparticles to act as a vaccine adjuvant. TNF/CHP nanoparticles showed excellent storage stability and enhanced host immune responses to external immunogens. The nanoparticles were effective via the nasal route of administration for inducing systemic IgG1 as well as mucosal IgA. We applied the nanoparticles in a model experimental influenza virus infection to investigate their adjuvant ability. TNF/CHP nanoparticles combined with a conventional split vaccine protected mice via nasal administration against a lethal challenge of A/PR/8/34 (H1N1) influenza virus. Mechanistic studies showed that the nanoparticles enhanced antigen uptake by dendritic cells (DCs) and moderately induced the expression of inflammation-related genes in nasopharynx lymphoid tissue (NALT), leading to the activation of both B and T cells. Preliminary safety study revealed no severe toxicity to TNF/CHP nanoparticles. Slight-to-moderate influences in nasal mucosa were observed only in the repeated administration and they seemed to be reversible. Our data show that TNF/CHP nanoparticles effectively enhance both humoral and cellular immunity and could be a potential adjuvant for vaccines against infectious diseases, especially in the mucosa.
Collapse
|
18
|
Jarillo-Luna RA, Rivera-Aguilar V, Pacheco-Yépez J, Godínez-Victoria M, Oros-Pantoja R, Miliar-García A, Campos-Rodríguez R. Nasal IgA secretion in a murine model of acute stress. The possible role of catecholamines. J Neuroimmunol 2015; 278:223-31. [DOI: 10.1016/j.jneuroim.2014.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 10/14/2014] [Accepted: 11/09/2014] [Indexed: 01/23/2023]
|
19
|
Moreno-Fierros L, García-Hernández AL, Ilhuicatzi-Alvarado D, Rivera-Santiago L, Torres-Martínez M, Rubio-Infante N, Legorreta-Herrera M. Cry1Ac protoxin from Bacillus thuringiensis promotes macrophage activation by upregulating CD80 and CD86 and by inducing IL-6, MCP-1 and TNF-α cytokines. Int Immunopharmacol 2013; 17:1051-66. [PMID: 24157331 DOI: 10.1016/j.intimp.2013.10.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 10/03/2013] [Accepted: 10/04/2013] [Indexed: 01/17/2023]
Abstract
Bacillus thuringiensis Cry1Ac protoxin (pCry1Ac) is a promising mucosal adjuvant, but its action mechanism is unknown. We examined in vivo whether pCry1Ac promotes the activation of macrophages in the peritoneum, spleen and mesenteric lymph nodes or in the lungs and bronchoalveolar lavage after intraperitoneal or intranasal pCry1Ac administration, respectively, in BALB/c mice. pCry1Ac upregulated the costimulatory molecules CD80 and CD86 in these macrophages, but with distinct kinetics. In vitro stimulation of resident macrophages with pCry1Ac upregulated CD80 and CD86 and enhanced the production of the pro-inflammatory cytokines TNF-α, IL-6 and MCP-1. To investigate whether the pCry1Ac-induced activation was mediated through MAPK pathways, we pretreated RAW 264.7 cells with signaling inhibitors of MEK, JNK and p38 MAPKs (PD98059, SP600125 and SB203580, respectively). pCry1Ac-induced upregulation of CD86 and CD80 was partially inhibited by the MEK inhibitor. While LPS-induced upregulation mechanisms of CD80 and CD86 appear to be different; as these were particularly inhibited by MEK and JNK inhibitors, respectively. pCry1Ac-induced IL-6 and MCP-1 production was especially inhibited with the p38 MAPK inhibitor, whereas TNF-α was only slightly inhibited upon treatment with JNK and p38 MAPK inhibitors. Therefore macrophage stimulation with pCry1Ac induced the upregulation of CD80 and CD86, and the production of IL-6, TNF-α and MCP-1, possibly, through the MEK and p38 MAPK pathways. It also promoted the nuclear translocation of NF-κB p50 and p65, the upregulation of MHC-II, and the activation of T CD4+ cells. These results suggest that pCry1Ac induced macrophage activation through mechanisms which differ partially from the LPS-induced.
Collapse
Affiliation(s)
- Leticia Moreno-Fierros
- Laboratorio de Inmunidad en Mucosas, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla 54090, Estado de México, México.
| | | | | | | | | | | | | |
Collapse
|
20
|
PLGA, PLGA-TMC and TMC-TPP nanoparticles differentially modulate the outcome of nasal vaccination by inducing tolerance or enhancing humoral immunity. PLoS One 2011; 6:e26684. [PMID: 22073184 PMCID: PMC3206834 DOI: 10.1371/journal.pone.0026684] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Accepted: 10/02/2011] [Indexed: 01/04/2023] Open
Abstract
Development of vaccines in autoimmune diseases has received wide attention over the last decade. However, many vaccines showed limited clinical efficacy. To enhance vaccine efficacy in infectious diseases, biocompatible and biodegradable polymeric nanoparticles have gained interest as antigen delivery systems. We investigated in mice whether antigen-encapsulated PLGA (poly-lactic-co-glycolic acid), PLGA-TMC (N-trimethyl chitosan) or TMC-TPP (tri-polyphosphate) nanoparticles can also be used to modulate the immunological outcome after nasal vaccination. These three nanoparticles enhanced the antigen presentation by dendritic cells, as shown by increased in vitro and in vivo CD4+ T-cell proliferation. However, only nasal PLGA nanoparticles were found to induce an immunoregulatory response as shown by enhanced Foxp3 expression in the nasopharynx associated lymphoid tissue and cervical lymph nodes. Nasal administration of OVA-containing PLGA particle resulted in functional suppression of an OVA-specific Th-1 mediated delayed-type hypersensitivity reaction, while TMC-TPP nanoparticles induced humoral immunity, which coincided with the enhanced generation of OVA-specific B-cells in the cervical lymph nodes. Intranasal treatment with Hsp70-mB29a peptide-loaded PLGA nanoparticles suppressed proteoglycan-induced arthritis, leading to a significant reduction of disease. We have uncovered a role for PLGA nanoparticles to enhance CD4+ T-cell mediated immunomodulation after nasal application. The exploitation of this differential regulation of nanoparticles to modulate nasal immune responses can lead to innovative vaccine development for prophylactic or therapeutic vaccination in infectious or autoimmune diseases.
Collapse
|
21
|
Differences between the ovine tonsils based on an immunohistochemical quantification of the lymphocyte subpopulations. Comp Immunol Microbiol Infect Dis 2011; 34:217-25. [DOI: 10.1016/j.cimid.2010.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 11/10/2010] [Indexed: 11/22/2022]
|
22
|
Oros-Pantoja R, Jarillo-Luna A, Rivera-Aguilar V, Sánchez-Torres LE, Godinez-Victoria M, Campos-Rodríguez R. Effects of restraint stress on NALT structure and nasal IgA levels. Immunol Lett 2010; 135:78-87. [PMID: 20937309 DOI: 10.1016/j.imlet.2010.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2010] [Revised: 09/23/2010] [Accepted: 10/03/2010] [Indexed: 02/02/2023]
Abstract
The effects of stress on the mucosal immune responses in inflammatory disorders of the gut, as well as on salivary and intestinal IgA levels are well known. However, its effects on the structure and function of the NALT have not yet been reported, and are examined in the present study. Balb/c mice were submitted to restraint stress for 3h per day during 4 or 8d. The immunohistochemistry and flow cytometric analysis revealed that repeated restraint stress (4 and 8d) decreased the percentage, compared to the control group, of CD3(+) and CD4(+) T cells, without affecting the percentage of CD8(+) T cells or B220(+) cells (B cells). The numbers of IELs (CD4(+) and CD8(+) T cells) were lower at 4d of stress and higher at 8d. IgA(+) cells in NALT and nasal IgA levels showed a similar pattern, being significantly lower at 4d of stress and significantly higher at 8d. In summary, repeated restraint stress altered the distribution and number of lymphocytes and IgA(+) cells in nasal mucosa, probably due to changes in norepinephrine and corticosterone levels.
Collapse
Affiliation(s)
- Rigoberto Oros-Pantoja
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Miron, CP. 11340, México, DF, Mexico
| | | | | | | | | | | |
Collapse
|
23
|
Rodriguez-Monroy MA, Moreno-Fierros L. Striking activation of NALT and nasal passages lymphocytes induced by intranasal immunization with Cry1Ac protoxin. Scand J Immunol 2010; 71:159-68. [PMID: 20415781 DOI: 10.1111/j.1365-3083.2009.02358.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cry1Ac protoxin from Bacillus thuringiensis is a potent mucosal immunogen and adjuvant. When delivered intranasally (i.n.) Cry1Ac elicits significant antibody response and is able to improve vaccination against Naegleria fowleri infection, but the functional effects occurring in nasal lymphocytes when this protein is administered alone have not been determined. Here, we investigated the effects of i.n. immunization with Cry1Ac on antibody production, lymphocyte activation and cytokine production in lymphocytes from nasal-associated lymphoid tissue (NALT) and nasal passages (NP). Our results show that i.n. immunization with Cry1Ac induced significant specific IgA and IgG cell responses, especially in NP. Besides, it increased the proportion of lymphocytes expressing the activation markers CD25 and CD69 in both nasal tissues, but differently. CD25 was increased in B cells along with CD4 and CD8 T cells from NALT and NP, while CD69 was increased in B cells from both tissues but only in CD4 T cells from NP. Finally, we found that Cry1Ac augmented especially a Th2 profile of cytokines, as the proportion of T cells that spontaneously produced IL-4, IL-5 and IL-10 was increased and this effect was higher in NP than in NALT. These data contribute to explain the potent immunogenicity of Cry1Ac via i.n. route.
Collapse
Affiliation(s)
- M A Rodriguez-Monroy
- Carrera de Médico Cirujano, Unidad de Biomedicina F.E.S. Iztacala, Universidad Nacional Autónoma de México, Tlalnepantia, Edo. México, México
| | | |
Collapse
|
24
|
Zygmunt BM, Rharbaoui F, Groebe L, Guzman CA. Intranasal Immunization Promotes Th17 Immune Responses. THE JOURNAL OF IMMUNOLOGY 2009; 183:6933-8. [DOI: 10.4049/jimmunol.0901144] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
25
|
Wissinger EL, Saldana J, Didierlaurent A, Hussell T. Manipulation of acute inflammatory lung disease. Mucosal Immunol 2008; 1:265-78. [PMID: 19079188 PMCID: PMC7100270 DOI: 10.1038/mi.2008.16] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Accepted: 02/26/2008] [Indexed: 02/04/2023]
Abstract
Inflammatory lung disease to innocuous antigens or infectious pathogens is a common occurrence and in some cases, life threatening. Often, the inflammatory infiltrate that accompanies these events contributes to pathology by deleterious effects on otherwise healthy tissue and by compromising lung function by consolidating (blocking) the airspaces. A fine balance, therefore, exists between a lung immune response and immune-mediated damage, and in some the "threshold of ignorance" may be set too low. In most cases, the contributing, potentially offending, cell population or immune pathway is known, as are factors that regulate them. Why then are targeted therapeutic strategies to manipulate them not more commonplace in clinical medicine? This review highlights immune homeostasis in the lung, how and why this is lost during acute lung infection, and strategies showing promise as future immune therapeutics.
Collapse
Affiliation(s)
- E L Wissinger
- Imperial College London, Kennedy Institute of Rheumatology, London, UK
| | - J Saldana
- Imperial College London, Kennedy Institute of Rheumatology, London, UK
| | - A Didierlaurent
- Imperial College London, Kennedy Institute of Rheumatology, London, UK
- Present Address: Present address: GlaxoSmithKline Biologicals, Rue de l'Institut 89, Rixensart B-1330, Belgium,
| | - T Hussell
- Imperial College London, Kennedy Institute of Rheumatology, London, UK
| |
Collapse
|