1
|
Napiórkowska-Baran K, Darwish S, Kaczor J, Treichel P, Szymczak B, Szota M, Koperska K, Bartuzi Z. Oral Diseases as a Manifestation of Inborn Errors of Immunity. J Clin Med 2024; 13:5079. [PMID: 39274292 PMCID: PMC11396297 DOI: 10.3390/jcm13175079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
Oral findings such as inflammation, ulcerations, or lesions can indicate serious systemic diseases and should prompt suspicion of acquired chronic conditions or inborn errors of immunity (IEIs). Currently, there are approximately 500 disease entities classified as IEIs, with the list expanding annually. The awareness of the existence of such conditions is of paramount importance, as patients with these disorders frequently necessitate the utilization of enhanced diagnostic techniques. This is exemplified by patients with impaired antibody production, in whom conventional serological methods may prove to be undiagnostic. Patients with IEI may require distinct therapeutic approaches or antimicrobial prophylaxis throughout their lives. An accurate diagnosis and, more importantly, early identification of patients with immune deficiencies is crucial to ensure the quality and longevity of their lives. It is important to note that the failure to establish a proper diagnosis or to provide adequate treatment could also have legal implications for medical professionals. The article presents IEIs, which may manifest in the oral cavity, and their diagnosis alongside therapeutic procedures.
Collapse
Affiliation(s)
- Katarzyna Napiórkowska-Baran
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Samira Darwish
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Justyna Kaczor
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Paweł Treichel
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Bartłomiej Szymczak
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Maciej Szota
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Kinga Koperska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| | - Zbigniew Bartuzi
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland
| |
Collapse
|
2
|
Li J, Xing H, Meng F, Liu T, Hong X, Han X, Dong Y, Li M, Wang Z, Zhang S, Cui C, Zheng A. Virus-Mimetic Extracellular-Vesicle Vaccine Boosts Systemic and Mucosal Immunity via Immune Recruitment. ACS NANO 2024. [PMID: 39013102 DOI: 10.1021/acsnano.4c01277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Mucosal vaccines can prevent viruses from infecting the respiratory mucosa, rather than only curtailing infection and protecting against the development of disease symptoms. The SARS-CoV-2 spike receptor-binding domain (RBD) is a compelling vaccine target but is undermined by suboptimal mucosal immunogenicity. Here, we report a SARS-CoV-2-mimetic extracellular-vesicle vaccine developed using genetic engineering and dendritic cell membrane budding. After mucosal immunization, the vaccine recruits antigen-presenting cells rapidly initiating a strong innate immune response. Notably, it obviates the need for adjuvants and can induce germinal center formation through both intramuscular and intratracheal vaccination. It not only elicits high levels of RBD-specific antibodies but also stimulates extensive cellular immunity in the respiratory mucosa. A sequential immunization strategy, starting with an intramuscular injection followed by an intratracheal booster, significantly bolsters mucosal immunity with high levels of IgA and tissue-resident memory T cell responses, thereby establishing a formidable defense against pseudovirus infection.
Collapse
Affiliation(s)
- Jingru Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Haonan Xing
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Fan Meng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Ting Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Xiaoxuan Hong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Xiaolu Han
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Yuhan Dong
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Meng Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zengming Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Shuang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Chunying Cui
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Capital Medical University, Beijing 100069, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing 100069, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing 100069, China
- Beijing Laboratory of Biomedical Materials, Beijing 100069, China
| | - Aiping Zheng
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
3
|
Sallard E, Aydin M. Cutting-edge research frontiers in oral cavity vaccines for respiratory diseases: a roadmap for scientific advancement. Front Cell Infect Microbiol 2024; 14:1388222. [PMID: 38988815 PMCID: PMC11234472 DOI: 10.3389/fcimb.2024.1388222] [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: 02/19/2024] [Accepted: 05/13/2024] [Indexed: 07/12/2024] Open
Abstract
Intramuscular vaccines present limitations in eliciting robust mucosal immunity and preventing respiratory pathogens transmission. Sublingual vaccine administration offers promising advantages, including interconnected mucosal protection. Despite these advantages, only a few clinical trials have explored sublingual vaccines, underscoring the necessity of optimizing next-generation vaccine formulas. Critical research priorities include understanding vector behavior in the oral environment, understanding their interactions with mucosal immunity and developing formulations enabling sustained mucosal contact to facilitate efficient transduction. Consequently, tonsil organoids, as representative human mucosal models, could offer critical insights into sublingual immunization. Thus, a multi-disciplinary approach integrating pharmacological, immunological, and manufacturing considerations is pivotal for sublingual vaccines in targeting pathogen-aggravated prevalent respiratory diseases including asthma, COPD and lung cancer, as well as the antimicrobial resistance crisis.
Collapse
Affiliation(s)
- Erwan Sallard
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
| | - Malik Aydin
- Virology and Microbiology, Center for Biomedical Education and Research (ZBAF), School of Medicine, Faculty of Health, Witten/Herdecke University, Witten, Germany
- Laboratory of Experimental Pediatric Pneumology and Allergology, Center for Biomedical Education and Research, School of Life Sciences (ZBAF), Faculty of Health, Witten/Herdecke University, Witten, Germany
- Institute for Medical Laboratory Diagnostics, Center for Clinical and Translational Research, Helios University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany
| |
Collapse
|
4
|
Ma B, Tao M, Li Z, Zheng Q, Wu H, Chen P. Mucosal vaccines for viral diseases: Status and prospects. Virology 2024; 593:110026. [PMID: 38373360 DOI: 10.1016/j.virol.2024.110026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Virus-associated infectious diseases are highly detrimental to human health and animal husbandry. Among all countermeasures against infectious diseases, prophylactic vaccines, which developed through traditional or novel approaches, offer potential benefits. More recently, mucosal vaccines attract attention for their extraordinary characteristics compared to conventional parenteral vaccines, particularly for mucosal-related pathogens. Representatively, coronavirus disease 2019 (COVID-19), a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), further accelerated the research and development efforts for mucosal vaccines by thoroughly investigating existing strategies or involving novel techniques. While several vaccine candidates achieved positive progresses, thus far, part of the current COVID-19 mucosal vaccines have shown poor performance, which underline the need for next-generation mucosal vaccines and corresponding platforms. In this review, we summarized the typical mucosal vaccines approved for humans or animals and sought to elucidate the underlying mechanisms of these successful cases. In addition, mucosal vaccines against COVID-19 that are in human clinical trials were reviewed in detail since this public health event mobilized all advanced technologies for possible solutions. Finally, the gaps in developing mucosal vaccines, potential solutions and prospects were discussed. Overall, rational application of mucosal vaccines would facilitate the establishing of mucosal immunity and block the transmission of viral diseases.
Collapse
Affiliation(s)
- Bingjie Ma
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Mengxiao Tao
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Zhili Li
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Quanfang Zheng
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Haigang Wu
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Peirong Chen
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China.
| |
Collapse
|
5
|
Feng X, Shi Y, Zhang Y, Lei F, Ren R, Tang X. Opportunities and Challenges for Inhalable Nanomedicine Formulations in Respiratory Diseases: A Review. Int J Nanomedicine 2024; 19:1509-1538. [PMID: 38384321 PMCID: PMC10880554 DOI: 10.2147/ijn.s446919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/24/2024] [Indexed: 02/23/2024] Open
Abstract
Lungs experience frequent interactions with the external environment and have an abundant supply of blood; therefore, they are susceptible to invasion by pathogenic microorganisms and tumor cells. However, the limited pharmacokinetics of conventional drugs in the lungs poses a clinical challenge. The emergence of different nano-formulations has been facilitated by advancements in nanotechnology. Inhaled nanomedicines exhibit better targeting and prolonged therapeutic effects. Although nano-formulations have great potential, they still present several unknown risks. Herein, we review the (1) physiological anatomy of the lungs and their biological barriers, (2) pharmacokinetics and toxicology of nanomaterial formulations in the lungs; (3) current nanomaterials that can be applied to the respiratory system and related design strategies, and (4) current applications of inhaled nanomaterials in treating respiratory disorders, vaccine design, and imaging detection based on the characteristics of different nanomaterials. Finally, (5) we analyze and summarize the challenges and prospects of nanomaterials for respiratory disease applications. We believe that nanomaterials, particularly inhaled nano-formulations, have excellent prospects for application in respiratory diseases. However, we emphasize that the simultaneous toxic side effects of biological nanomaterials must be considered during the application of these emerging medicines. This study aims to offer comprehensive guidelines and valuable insights for conducting research on nanomaterials in the domain of the respiratory system.
Collapse
Affiliation(s)
- Xujun Feng
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yuan Shi
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Ye Zhang
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Fei Lei
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Rong Ren
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Xiangdong Tang
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
6
|
Simões R, Ribeiro AC, Dias R, Freitas V, Soares S, Pérez-Gregorio R. Unveiling the Immunomodulatory Potential of Phenolic Compounds in Food Allergies. Nutrients 2024; 16:551. [PMID: 38398875 PMCID: PMC10891931 DOI: 10.3390/nu16040551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Food allergies are becoming ever more prevalent around the world. This pathology is characterized by the breakdown of oral tolerance to ingested food allergens, resulting in allergic reactions in subsequent exposures. Due to the possible severity of the symptoms associated with this pathology, new approaches to prevent it and reduce associated symptoms are of utmost importance. In this framework, dietary phenolic compounds appear as a tool with a not fully explored potential. Some phenolic compounds have been pointed to with the ability to modulate food allergies and possibly reduce their symptoms. These compounds can modulate food allergies through many different mechanisms, such as altering the bioaccessibility and bioavailability of potentially immunogenic peptides, by modulating the human immune system and by modulating the composition of the human microbiome that resides in the oral cavity and the gastrointestinal tract. This review deepens the state-of-the-art of the modulation of these mechanisms by phenolic compounds. While this review shows clear evidence that dietary supplementation with foods rich in phenolic compounds might constitute a new approach to the management of food allergies, it also highlights the need for further research to delve into the mechanisms of action of these compounds and decipher systematic structure/activity relationships.
Collapse
Affiliation(s)
- Rodolfo Simões
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
- Food and Health Omics Group, Food and Agroecology Institute, University of Vigo, Campus As Lagoas, s/n, 32004 Ourense, Spain
- Food and Health Omics Group, Department of Chemistry and Biochemistry, Galicia Sur Health Research Institute (IISGS), SERGAS-UVIGO, 32002 Ourense, Spain
| | - Ana Catarina Ribeiro
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
| | - Ricardo Dias
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
| | - Victor Freitas
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
| | - Susana Soares
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
| | - Rosa Pérez-Gregorio
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua Campo Alegre 687, s/n, 4169-007 Porto, Portugal
- Food and Health Omics Group, Food and Agroecology Institute, University of Vigo, Campus As Lagoas, s/n, 32004 Ourense, Spain
- Food and Health Omics Group, Department of Chemistry and Biochemistry, Galicia Sur Health Research Institute (IISGS), SERGAS-UVIGO, 32002 Ourense, Spain
| |
Collapse
|
7
|
Zhang C, Zhao Z, Jia YJ, Zhang PQ, Sun Y, Zhou YC, Wang GX, Zhu B. Rationally Designed Self-Assembling Nanovaccines Elicit Robust Mucosal and Systemic Immunity against Rhabdovirus. ACS APPLIED MATERIALS & INTERFACES 2024; 16:228-244. [PMID: 38055273 DOI: 10.1021/acsami.3c14305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Viral diseases have constantly caused great threats to global public health, resulting in an urgent need for effective vaccines. However, the current viral vaccines often show low immunogenicity. To counter this, we report a smart strategy of a well-designed modular nanoparticle (LSG-TDH) that recapitulates the dominant antigen SG, low-molecular-weight protamine, and tetralysine-modified H-chain apoferritin (TDH). The constructed LSG-TDH nanovaccine could self-assemble into a nanocage structure, which confers excellent mucus-penetrating, cellular affinity, and uptake ability. Studies demonstrate that the LSG-TDH nanovaccine could strongly activate both mucosal and systemic immune responses. Importantly, by immunizing wild-type and TLR2 knockout (TLR2-KO) zebrafish, we found that TLR2 could mediate LSG-TDH-induced adaptive mucosal and systemic immune responses by activating antigen-presenting cells. Collectively, our findings offer new insights into rational viral vaccine design and provide additional evidence of the vital role of TLR2 in regulating adaptive immunity.
Collapse
Affiliation(s)
- Chen Zhang
- Collaborative Innovation Center of Marine Science and Technology, Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou 570228, P. R. China
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Zhao Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Yi-Jun Jia
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Peng-Qi Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Yun Sun
- Collaborative Innovation Center of Marine Science and Technology, Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou 570228, P. R. China
| | - Yong-Can Zhou
- Collaborative Innovation Center of Marine Science and Technology, Hainan Provincial Key Laboratory for Tropical Hydrobiology and Biotechnology, College of Marine Science, Hainan University, Haikou 570228, P. R. China
| | - Gao-Xue Wang
- College of Animal Science and Technology, Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| | - Bin Zhu
- College of Animal Science and Technology, Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, P. R. China
| |
Collapse
|
8
|
Rao Z, Zhou H, Li Q, Zeng N, Wang Q. Extraction, purification, structural characteristics and biological properties of the polysaccharides from Radix Saposhnikoviae: A review. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116956. [PMID: 37487960 DOI: 10.1016/j.jep.2023.116956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/09/2023] [Accepted: 07/21/2023] [Indexed: 07/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix Saposhnikoviae (R. Saposhnikoviae), commonly known as FangFeng, is a renowned medicinal herb in China extensively utilized in traditional Chinese medicine. It expels pathogenic wind from the body surface, alleviates pain by removing dampness, and relieves convulsion. Therefore, it is mainly used for treating exterior syndrome, itchy wind papules, rheumatic arthralgia, and splenic asthenia-induced dampness. R. saposhnikoviae has important medicinal value, and the polysaccharide component is one of its important active ingredients. AIM OF THE REVIEW This review summarizes the factors influencing the content of polysaccharides in R. Saposhnikoviae (PRSs), the techniques employed for their extraction, separation, and purification, their structural characterization, and their biological activities. MATERIALS AND METHODS Relevant research reports on PRSs were collected from the Chinese National Knowledge Infrastructure, Web of Science, PubMed, Wanfang Data Knowledge Service Platform, China Master Theses Full-text Database, and China Doctoral Dissertations Full-text Database. RESULTS The content of PRSs can vary depending on cultivation methods and harvesting time. PRSs were extracted using various extraction techniques such as hot water, ultrasonic-assisted, microwave-assisted, and enzymatic extractions, as well as water extraction and alcohol precipitation. Effective purification methods involve protein removal using trifluoro-trichloroethane and the decolorization of the polysaccharide using column chromatography with D280 anion exchange resins. Current research highlights the significant pharmacological activities of PRSs in R. Saposhnikoviae, including immunomodulatory, antioxidant, anti-allergic, anti-cancer, and anti-osteoporotic effects as well as prevention of calcium loss and maintenance of mucosal function. CONCLUSIONS PRSs play a crucial role as bioactive constituents of R. Saposhnikoviae, exhibiting diverse biological activities and promising applications. A deeper understanding of PRSs will contribute to the improved utilization of R. Saposhnikoviae and the development of related derivatives of the active ingredients.
Collapse
Affiliation(s)
- Zhili Rao
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine/The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, 400021, PR China; Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, PR China
| | - Hongli Zhou
- National Drug Clinical Trial Institution, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, PR China
| | - Qian Li
- Rehabilitation Department, Chongqing Hospital of Traditional Chinese Medicine/The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, 400021, PR China
| | - Nan Zeng
- Department of Pharmacology, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, PR China.
| | - Qin Wang
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine/The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, 400021, PR China.
| |
Collapse
|
9
|
Liu Y, Dong Y, Shen W, DU J, Sun Q, Yang Y, Yin D. Platycodon grandiflorus polysaccharide regulates colonic immunity through mesenteric lymphatic circulation to attenuate ulcerative colitis. Chin J Nat Med 2023; 21:263-278. [PMID: 37120245 DOI: 10.1016/s1875-5364(23)60435-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Indexed: 05/01/2023]
Abstract
Platycodon grandiflorus polysaccharide (PGP) is one of the main components of P. grandiflorus, but the mechanism of its anti-inflammatory effect has not been fully elucidated. The aim of this study was to evaluate the therapeutic effect of PGP on mice with dextran sodium sulfate (DSS)-induced ulcerative colitis (UC) and explore the underlying mechanisms. The results showed that PGP treatment inhibited the weight loss of DSS-induced UC mice, increased colon length, and reduced DAI, spleen index, and pathological damage within the colon. PGP also reduced the levels of pro-inflammatory cytokines and inhibited the enhancement of oxidative stress and MPO activity. Meanwhile, PGP restored the levels of Th1, Th2, Th17, and Treg cell-related cytokines and transcription factors in the colon to regulate colonic immunity. Further studies revealed that PGP regulated the balance of colonic immune cells through mesenteric lymphatic circulation. Taken together, PGP exerts anti-inflammatory and anti-oxidant effect and regulates colonic immunity to attenuate DSS-induced UC through mesenteric lymphatic circulation.
Collapse
Affiliation(s)
- Yang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yahui Dong
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China
| | - Jiahui DU
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Anhui Provincial Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230021, China.
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China; Anhui Provincial Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230021, China; Anhui Provincial Key Laboratory of Research & Development of Chinese Medicine, Hefei 230021, China.
| |
Collapse
|
10
|
Nakahashi-Ouchida R, Fujihashi K, Kurashima Y, Yuki Y, Kiyono H. Nasal vaccines: solutions for respiratory infectious diseases. Trends Mol Med 2023; 29:124-140. [PMID: 36435633 DOI: 10.1016/j.molmed.2022.10.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/07/2022] [Accepted: 10/26/2022] [Indexed: 11/25/2022]
Abstract
Nasal vaccines induce pathogen-specific dual protective immunity at mucosal surfaces and systemically throughout the body. Consequently, nasal vaccines both prevent pathogen invasion and reduce disease severity. Because of these features, nasal vaccines are considered to be a next-generation tool for preventing respiratory infectious diseases, including COVID-19. However, nasal vaccines must overcome key safety concerns given the anatomic proximity of the central nervous system (CNS) via the olfactory bulbs which lie next to the nasal cavity. This review summarizes current efforts to develop safe and effective nasal vaccines and delivery systems, as well as their clinical applications for the prevention of respiratory infections. We also discuss various concerns regarding the safety of nasal vaccines and introduce a system for evaluating them.
Collapse
Affiliation(s)
- Rika Nakahashi-Ouchida
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan; Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yosuke Kurashima
- Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan; Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, Chiba, Japan; Chiba University-University of California San Diego (CU-UCSD) Center for Mucosal Immunology, Allergy, and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
| | - Yoshikazu Yuki
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; HanaVax Inc., Tokyo, Japan
| | - Hiroshi Kiyono
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan; Research Institute of Disaster Medicine, Chiba University, Chiba, Japan; Future Mucosal Vaccine Research and Development Synergy Institute, Chiba University, Chiba, Japan; Institute for Advanced Academic Research, Chiba University, Chiba, Japan; Chiba University-University of California San Diego (CU-UCSD) Center for Mucosal Immunology, Allergy, and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA; Future Medicine Education and Research Organization, Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan.
| |
Collapse
|
11
|
Moreno CM, Boeree E, Freitas CMT, Weber KS. Immunomodulatory role of oral microbiota in inflammatory diseases and allergic conditions. FRONTIERS IN ALLERGY 2023; 4:1067483. [PMID: 36873050 PMCID: PMC9981797 DOI: 10.3389/falgy.2023.1067483] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/26/2023] [Indexed: 02/19/2023] Open
Abstract
In recent years, the interplay between oral microbiota and systemic disease has gained attention as poor oral health is associated with several pathologies. The oral microbiota plays a role in the maintenance of overall health, and its dysbiosis influences chronic inflammation and the pathogenesis of gum diseases. Periodontitis has also been associated with other diseases and health complications such as cancer, neurogenerative and autoimmune disorders, chronic kidney disease, cardiovascular diseases, rheumatic arthritis, respiratory health, and adverse pregnancy outcomes. The host microbiota can influence immune cell development and immune responses, and recent evidence suggests that changes in oral microbiota composition may also contribute to sensitization and the development of allergic reactions, including asthma and peanut allergies. Conversely, there is also evidence that allergic reactions within the gut may contribute to alterations in oral microbiota composition. Here we review the current evidence of the role of the oral microbiota in inflammatory diseases and health complications, as well as its future relevance in improving health and ameliorating allergic disease.
Collapse
Affiliation(s)
- Carlos M Moreno
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| | - Ellie Boeree
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| | - Claudia M Tellez Freitas
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, United States
| | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, United States
| |
Collapse
|
12
|
Tsai CJY, Loh JMS, Fujihashi K, Kiyono H. Mucosal vaccination: onward and upward. Expert Rev Vaccines 2023; 22:885-899. [PMID: 37817433 DOI: 10.1080/14760584.2023.2268724] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION The unique mucosal immune system allows the generation of robust protective immune responses at the front line of pathogen encounters. The needle-free delivery route and cold chain-free logistic requirements also provide additional advantages in ease and economy. However, the development of mucosal vaccines faces several challenges, and only a handful of mucosal vaccines are currently licensed. These vaccines are all in the form of live attenuated or inactivated whole organisms, whereas no subunit-based mucosal vaccine is available. AREAS COVERED The selection of antigen, delivery vehicle, route and adjuvants for mucosal vaccination are highly important. This is particularly crucial for subunit vaccines, as they often fail to elicit strong immune responses. Emerging research is providing new insights into the biological and immunological uniqueness of mucosal tissues. However, many aspects of the mucosal immunology still await to be investigated. EXPERT OPINION This article provides an overview of the current understanding of mucosal vaccination and discusses the remaining knowledge gaps. We emphasize that because of the potential benefits mucosal vaccines can bring from the biomedical, social and economic standpoints, the unmet goal to achieve mucosal vaccine success is worth the effort.
Collapse
Affiliation(s)
- Catherine J Y Tsai
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
| | - Jacelyn M S Loh
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, New Zealand, Auckland
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hiroshi Kiyono
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
- CU-UCSD Center for Mucosal Immunology, Allergy and Vaccines (cMAV), Division of Gastroenterology, Department of Medicine, University of California, San Diego, CA, USA
- Future Medicine Education and Research Organization, Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan
| |
Collapse
|
13
|
Zhao Z, Wang H, Zhang D, Guan Y, Siddiqui SA, Feng-Shan X, Cong B. Oral vaccination with recombinant Lactobacillus casei expressing Aeromonas hydrophila Aha1 against A. hydrophila infections in common carps. Virulence 2022; 13:794-807. [PMID: 35499101 PMCID: PMC9067532 DOI: 10.1080/21505594.2022.2063484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 11/03/2022] Open
Abstract
The immunogenicity of Aha1, an OMP of Aeromonas hydrophila mediating the adhesion of bacteria onto the mucosal surface of hosts has been established. In this study, recombinant vectors, pPG1 and pPG2, carrying a 1366 bp DNA fragment that was responsible for encoding the 49 kDa Aha1 from A. hydrophila were constructed, respectively, then electroporated into a probiotic strain Lactobacillus casei CC16 separately to generate two types of recombinants, L. casei-pPG1-Aha1 (Lc-pPG1-Aha1) and L. casei-pPG2-Aha1 (Lc-pPG2-Aha1). Subsequently, these were orally administered into common carps to examine their immunogenicity. The expression and localization of the expressed Aha1 protein relative to the carrier L. casei was validated via Western blotting, flow cytometry, and immune fluorescence separately. The recombinant vaccines produced were shown high efficacies, stimulated higher level of antibodies and AKP, ACP, SOD, LZM, C3, C4 in serum in hosts. Immune-related gene expressions of cytokines including IL-10, IL-1β, TNF-α, IFN-γ in the livers, spleens, HK, and intestines were up-regulated significantly. Besides, a more potent phagocytosis response was observed in immunized fish, and higher survival rates were presented in common carps immunized with Lc-pPG1-Aha1 (60%) and Lc-pPG2-Aha1 (50%) after re-infection with virulent strain A. hydrophila. Moreover, the recombinant L. casei were shown a stronger propensity for survivability in the intestine in immunized fish. Taken together, the recombinant L. casei strains might be promising candidates for oral vaccination against A. hydrophila infections in common carps.
Collapse
Affiliation(s)
- Zelin Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Hong Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Dongxing Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Yongchao Guan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Shahrood Ahmad Siddiqui
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Xiao Feng-Shan
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin, China
| | - Bo Cong
- Institute of special animal and plant sciences of CAAS, Changchun, Jilin, China
| |
Collapse
|
14
|
Liu Y, Chen Q, Ren R, Zhang Q, Yan G, Yin D, Zhang M, Yang Y. Platycodon grandiflorus polysaccharides deeply participate in the anti-chronic bronchitis effects of platycodon grandiflorus decoction, a representative of “the lung and intestine are related”. Front Pharmacol 2022; 13:927384. [PMID: 36160385 PMCID: PMC9489837 DOI: 10.3389/fphar.2022.927384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 07/22/2022] [Indexed: 11/22/2022] Open
Abstract
Platycodon grandiflorus (Jacq.) A. DC. (PG) root is one of the most commonly used medicine-food materials for respiratory discomfort in Asia, usually in the form of a decoction or leaching solution. As everyone knows, both of decoction and leaching solution is a polyphase dispersion system, containing low-molecular-weight water-soluble active ingredients and hydrophilic macromolecules. This study aimed to discuss the synergistic effect of Platycodon grandiflorus polysaccharide (PGP) and platycodin D (PD) in PG decoction against chronic bronchitis (CB) and the mechanism underlying. A series of PGP, PD, and PGD + PD suspensions were administrated to CB model rats, on the levels of whole animal and in situ intestinal segment with or without mesenteric lymphatic vessels ligation. It exhibited that PGP exhibited synergistic effects with PD, on improving the histopathological abnormity, mucus secretion excess, and immunological imbalance in lung of CB model rat, closely associated with its modulations on the mucosal immunity status in small intestine. The polysaccharide macromolecules in PG decoction or leaching solution should be responsible for the modulation of pulmonary immune state, possibly through the common mucosal immune between small intestine and lung. These results might be a new perspective that illustrates the classical theory of “the lung and intestine are related” in traditional Chinese medicine.
Collapse
Affiliation(s)
- Yang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qingqing Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Rongrong Ren
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qingqing Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Guiming Yan
- School of Nursing, Anhui University of Chinese Medicine, Hefei, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Anhui Provincial Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- *Correspondence: Dengke Yin, ; Ye Yang,
| | - Mingyan Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- School of Nursing, Anhui University of Chinese Medicine, Hefei, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- *Correspondence: Dengke Yin, ; Ye Yang,
| |
Collapse
|
15
|
Leong EWX, Ge R. Lipid Nanoparticles as Delivery Vehicles for Inhaled Therapeutics. Biomedicines 2022; 10:2179. [PMID: 36140280 PMCID: PMC9496059 DOI: 10.3390/biomedicines10092179] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Lipid nanoparticles (LNPs) have emerged as a powerful non-viral carrier for drug delivery. With the prevalence of respiratory diseases, particularly highlighted by the current COVID-19 pandemic, investigations into applying LNPs to deliver inhaled therapeutics directly to the lungs are underway. The progress in LNP development as well as the recent pre-clinical studies in three main classes of inhaled encapsulated drugs: small molecules, nucleic acids and proteins/peptides will be discussed. The advantages of the pulmonary drug delivery system such as reducing systemic toxicity and enabling higher local drug concentration in the lungs are evaluated together with the challenges and design considerations for improved formulations. This review provides a perspective on the future prospects of LNP-mediated delivery of inhaled therapeutics for respiratory diseases.
Collapse
Affiliation(s)
| | - Ruowen Ge
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117558, Singapore
| |
Collapse
|
16
|
Wang Z, Popowski KD, Zhu D, de Juan Abad BL, Wang X, Liu M, Lutz H, De Naeyer N, DeMarco CT, Denny TN, Dinh PUC, Li Z, Cheng K. Exosomes decorated with a recombinant SARS-CoV-2 receptor-binding domain as an inhalable COVID-19 vaccine. Nat Biomed Eng 2022; 6:791-805. [PMID: 35788687 PMCID: PMC10782831 DOI: 10.1038/s41551-022-00902-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 05/20/2022] [Indexed: 02/05/2023]
Abstract
The first two mRNA vaccines against infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that were approved by regulators require a cold chain and were designed to elicit systemic immunity via intramuscular injection. Here we report the design and preclinical testing of an inhalable virus-like-particle as a COVID-19 vaccine that, after lyophilisation, is stable at room temperature for over three months. The vaccine consists of a recombinant SARS-CoV-2 receptor-binding domain (RBD) conjugated to lung-derived exosomes which, with respect to liposomes, enhance the retention of the RBD in both the mucus-lined respiratory airway and in lung parenchyma. In mice, the vaccine elicited RBD-specific IgG antibodies, mucosal IgA responses and CD4+ and CD8+ T cells with a Th1-like cytokine expression profile in the animals' lungs, and cleared them of SARS-CoV-2 pseudovirus after a challenge. In hamsters, two doses of the vaccine attenuated severe pneumonia and reduced inflammatory infiltrates after a challenge with live SARS-CoV-2. Inhalable and room-temperature-stable virus-like particles may become promising vaccine candidates.
Collapse
Affiliation(s)
- Zhenzhen Wang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Kristen D Popowski
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | | | - Xianyun Wang
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Mengrui Liu
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA
| | - Halle Lutz
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Nicole De Naeyer
- Immunology and Virology Quality Assessment Center, Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - C Todd DeMarco
- Immunology and Virology Quality Assessment Center, Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Thomas N Denny
- Immunology and Virology Quality Assessment Center, Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Phuong-Uyen C Dinh
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA.
- Department of Pulmonary and Critical Care Medicine, Dongguan Institute of Respiratory and Critical Care Medicine, Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong, China.
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC, USA.
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, NC, USA.
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
17
|
Correa VA, Portilho AI, De Gaspari E. Vaccines, Adjuvants and Key Factors for Mucosal Immune Response. Immunology 2022; 167:124-138. [PMID: 35751397 DOI: 10.1111/imm.13526] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 04/26/2022] [Indexed: 11/29/2022] Open
Abstract
Vaccines are the most effective tool to control infectious diseases, which provoke significant morbidity and mortality. Most vaccines are administered through the parenteral route and can elicit a robust systemic humoral response, but they induce a weak T-cell-mediated immunity and are poor inducers of mucosal protection. Considering that most pathogens enter the body through mucosal surfaces, a vaccine that elicits protection in the first site of contact between the host and the pathogen is promising. However, despite the advantages of mucosal vaccines as good options to confer protection on the mucosal surface, only a few mucosal vaccines are currently approved. In this review, we discuss the impact of vaccine administration in different mucosal surfaces; how appropriate adjuvants enhance the induction of protective mucosal immunity and other factors that can influence the mucosal immune response to vaccines. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Victor Araujo Correa
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| | - Amanda Izeli Portilho
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| | - Elizabeth De Gaspari
- Adolfo Lutz Institute, Immunology Center, Av Dr Arnaldo, 355, 11th floor, room 1116, Cerqueira César, São Paulo, SP, Brazil.,São Paulo University, Biomedical Sciences Institute, Graduate Program Interunits in Biotechnology, Av Prof Lineu Prestes, 2415, ICB III, São Paulo, SP, Brazil
| |
Collapse
|
18
|
Barreto de Albuquerque J, Altenburger LM, Abe J, von Werdt D, Wissmann S, Martínez Magdaleno J, Francisco D, van Geest G, Ficht X, Iannacone M, Bruggmann R, Mueller C, Stein JV. Microbial uptake in oral mucosa-draining lymph nodes leads to rapid release of cytotoxic CD8 + T cells lacking a gut-homing phenotype. Sci Immunol 2022; 7:eabf1861. [PMID: 35714202 DOI: 10.1126/sciimmunol.abf1861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The gastrointestinal (GI) tract constitutes an essential barrier against ingested microbes, including potential pathogens. Although immune reactions are well studied in the lower GI tract, it remains unclear how adaptive immune responses are initiated during microbial challenge of the oral mucosa (OM), the primary site of microbial encounter in the upper GI tract. Here, we identify mandibular lymph nodes (mandLNs) as sentinel lymphoid organs that intercept ingested Listeria monocytogenes (Lm). Oral Lm uptake led to local activation and release of antigen-specific CD8+ T cells that constituted most of the early circulating effector T cell (TEFF) pool. MandLN-primed TEFF disseminated to lymphoid organs, lung, and OM and contributed substantially to rapid elimination of target cells. In contrast to CD8+ TEFF generated in mesenteric LN (MLN) during intragastric infection, mandLN-primed TEFF lacked a gut-seeking phenotype, which correlated with low expression of enzymes required for gut-homing imprinting by mandLN stromal and dendritic cells. Accordingly, mandLN-primed TEFF decreased Lm burden in spleen but not MLN after intestinal infection. Our findings extend the concept of regional specialization of immune responses along the length of the GI tract, with CD8+ TEFF generated in the upper GI tract displaying homing profiles that differ from those imprinted by lymphoid tissue of the lower GI tract.
Collapse
Affiliation(s)
| | - Lukas M Altenburger
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Jun Abe
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Diego von Werdt
- Division of Experimental Pathology, Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Stefanie Wissmann
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - Jose Martínez Magdaleno
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| | - David Francisco
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, 3012 Bern, Switzerland
| | - Geert van Geest
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, 3012 Bern, Switzerland
| | - Xenia Ficht
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy.,Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Remy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, 3012 Bern, Switzerland
| | - Christoph Mueller
- Division of Experimental Pathology, Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
19
|
Bergmeier LA, Dutzan N, Smith PC, Kraan H. Editorial: Immunology of the Oral Mucosa. Front Immunol 2022; 13:877209. [PMID: 35401502 PMCID: PMC8992007 DOI: 10.3389/fimmu.2022.877209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/04/2022] [Indexed: 12/03/2022] Open
Affiliation(s)
- Lesley Ann Bergmeier
- Centre for Immunobiology and Regenerative Medicine, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Nicolas Dutzan
- Department of Conservative Dentistry, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Patricio C Smith
- School of Dentistry, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Heleen Kraan
- Institute for Translational Vaccinology, Intravacc, Bilthoven, Netherlands
| |
Collapse
|
20
|
Trincado V, Gala RP, Morales JO. Buccal and Sublingual Vaccines: A Review on Oral Mucosal Immunization and Delivery Systems. Vaccines (Basel) 2021; 9:vaccines9101177. [PMID: 34696284 PMCID: PMC8539688 DOI: 10.3390/vaccines9101177] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, most vaccines available on the market are for parental use; however, this may not be the best option on several occasions. Mucosal routes of administration such as intranasal, sublingual, and buccal generate great interest due to the benefits they offer. These range from increasing patient compliance to inducing a more effective immune response than that achieved through conventional routes. Due to the activation of the common mucosal immune system, it is possible to generate an effective systemic and local immune response, which is not achieved through parenteral administration. Protection against pathogens that use mucosal entry routes is provided by an effective induction of mucosal immunity. Mucosal delivery systems are being developed, such as films and microneedles, which have proven to be effective, safe, and easy to administer. These systems have multiple advantages over commonly used injections, which are simple to manufacture, stable at room temperature, painless for the patient since they do not require puncture. Therefore, these delivery systems do not require to be administered by medical personnel; in fact, they could be self-administered.
Collapse
Affiliation(s)
- Valeria Trincado
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
| | - Rikhav P. Gala
- Biotechnology Division, Center Mid-Atlantic, Fraunhofer USA, Newark, DE 19702, USA;
| | - Javier O. Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
- Correspondence:
| |
Collapse
|
21
|
Bou Zerdan M, Moussa S, Atoui A, Assi HI. Mechanisms of Immunotoxicity: Stressors and Evaluators. Int J Mol Sci 2021; 22:8242. [PMID: 34361007 PMCID: PMC8348050 DOI: 10.3390/ijms22158242] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/12/2022] Open
Abstract
The immune system defends the body against certain tumor cells and against foreign agents such as fungi, parasites, bacteria, and viruses. One of its main roles is to distinguish endogenous components from non-self-components. An unproperly functioning immune system is prone to primary immune deficiencies caused by either primary immune deficiencies such as genetic defects or secondary immune deficiencies such as physical, chemical, and in some instances, psychological stressors. In the manuscript, we will provide a brief overview of the immune system and immunotoxicology. We will also describe the biochemical mechanisms of immunotoxicants and how to evaluate immunotoxicity.
Collapse
Affiliation(s)
- Maroun Bou Zerdan
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, 1107 2020 Beirut, Lebanon; (M.B.Z.); (A.A.)
| | - Sara Moussa
- Faculty of Medicine, University of Balamand, 1100 Beirut, Lebanon;
| | - Ali Atoui
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, 1107 2020 Beirut, Lebanon; (M.B.Z.); (A.A.)
| | - Hazem I. Assi
- Department of Internal Medicine, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, 1107 2020 Beirut, Lebanon; (M.B.Z.); (A.A.)
| |
Collapse
|
22
|
Cugini C, Ramasubbu N, Tsiagbe VK, Fine DH. Dysbiosis From a Microbial and Host Perspective Relative to Oral Health and Disease. Front Microbiol 2021; 12:617485. [PMID: 33763040 PMCID: PMC7982844 DOI: 10.3389/fmicb.2021.617485] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022] Open
Abstract
The significance of microbiology and immunology with regard to caries and periodontal disease gained substantial clinical or research consideration in the mid 1960's. This enhanced emphasis related to several simple but elegant experiments illustrating the relevance of bacteria to oral infections. Since that point, the understanding of oral diseases has become increasingly sophisticated and many of the original hypotheses related to disease causality have either been abandoned or amplified. The COVID pandemic has reminded us of the importance of history relative to infectious diseases and in the words of Churchill "those who fail to learn from history are condemned to repeat it." This review is designed to present an overview of broad general directions of research over the last 60 years in oral microbiology and immunology, reviewing significant contributions, indicating emerging foci of interest, and proposing future directions based on technical advances and new understandings. Our goal is to review this rich history (standard microbiology and immunology) and point to potential directions in the future (omics) that can lead to a better understanding of disease. Over the years, research scientists have moved from a position of downplaying the role of bacteria in oral disease to one implicating bacteria as true pathogens that cause disease. More recently it has been proposed that bacteria form the ecological first line of defense against "foreign" invaders and also serve to train the immune system as an acquired host defensive stimulus. While early immunological research was focused on immunological exposure as a modulator of disease, the "hygiene hypothesis," and now the "old friends hypothesis" suggest that the immune response could be trained by bacteria for long-term health. Advanced "omics" technologies are currently being used to address changes that occur in the host and the microbiome in oral disease. The "omics" methodologies have shaped the detection of quantifiable biomarkers to define human physiology and pathologies. In summary, this review will emphasize the role that commensals and pathobionts play in their interaction with the immune status of the host, with a prediction that current "omic" technologies will allow researchers to better understand disease in the future.
Collapse
Affiliation(s)
- Carla Cugini
- Department of Oral Biology, Rutgers School of Dental Medicine, Newark, NJ, United States
| | | | | | | |
Collapse
|
23
|
The Role of Mucosal Immunity and Recombinant Probiotics in SARS-CoV2 Vaccine Development. Probiotics Antimicrob Proteins 2021; 13:1239-1253. [PMID: 33770348 PMCID: PMC7996120 DOI: 10.1007/s12602-021-09773-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 01/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), causing the 2019 novel coronavirus disease (COVID-19), was introduced by WHO (World Health Organization) as "pandemic" in March 2020. According to WHO, thus far (23 November 2020) 58,425,681 infected cases including 1,385,218 deaths have been reported worldwide. In order to reduce transmission and spread of this lethal virus, attempts are globally being made to develop an appropriate vaccine. Intending to neutralize pathogens at their initial entrance site, protective mucosal immunity is inevitably required. In SARS-CoV2 infection and transmission, respiratory mucosa plays a key role; hence, apparently mucosal vaccination could be a superior approach to elicit mucosal and systemic immune responses simultaneously. In this review, the advantages of mucosal vaccination to control COVID-19 infection, limitations, and outcomes of mucosal vaccines have been highlighted. Considering the gut microbiota dysregulation in COVID-19, we further provide evidences on utilization of recombinant probiotics, particularly lactic acid bacteria (LAB) as vaccine carrier. Their intrinsic immunomodulatory features, natural adjuvanticity, and feasible expression of relevant antigen in the mucosal surface make them more appealing as live cell factory. Among all available platforms, bioengineered probiotics are considered as the most affordable, most practical, and safest vaccination approach to halt this emerging virus.
Collapse
|
24
|
Ryu B, Baek J, Kim H, Lee JH, Kim J, Jeong YH, Lee SG, Kang KR, Oh MS, Kim EY, Kim CY, Chung HM. Anti-Inflammatory Effects of M-MSCs in DNCB-Induced Atopic Dermatitis Mice. Biomedicines 2020; 8:biomedicines8100439. [PMID: 33096640 PMCID: PMC7589030 DOI: 10.3390/biomedicines8100439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/12/2020] [Accepted: 10/20/2020] [Indexed: 01/03/2023] Open
Abstract
Atopic dermatitis (AD) is an inflammatory skin disease caused by an imbalance between Th1 and Th2 cells. AD patients suffer from pruritus, excessive dryness, red or inflamed skin, and complications such as sleep disturbances and depression. Although there are currently many AD treatments available there are insufficient data on their long-term stability and comparative effects. Moreover, they have limitations due to various side effects. Multipotent mesenchymal stem cells (M-MSCs) might have potential for next-generation AD therapies. MSCs are capable of immune function regulation and local inflammatory response inhibition. M-MSCs, derived from human embryonic stem cells (hESC), additionally have a stable supply. In L507 antibody array, M-MSCs generally showed similar tendencies to bone marrow-derived mesenchymal stem cells (BM-MSCs), although the immunoregulatory function of M-MSCs seemed to be superior to BM-MSCs. Based on the characteristics of M-MSCs on immunoregulatory functions, we tested a M-MSC conditioned media concentrate (MCMC) in mice with AD lesions on their dorsal skin. MCMC significantly decreased RNA expression levels of inflammatory cytokines in the mouse dorsal skin. It also suppressed serum IgE levels. In addition, significant histopathologic alleviation was identified. In conclusion, secretions of M-MSCs have the potential to effectively improve AD-related inflammatory lesions. M-MSCs showed potential for use in next-generation AD treatment.
Collapse
Affiliation(s)
- Bokyeong Ryu
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (B.R.); (J.K.)
| | - Jieun Baek
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
| | - Hana Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
| | - Ji-Heon Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
| | - Jin Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (B.R.); (J.K.)
| | - Young-Hoon Jeong
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
| | - Seul-Gi Lee
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
| | - Kyu-Ree Kang
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
| | - Min-Seok Oh
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | | | - C-Yoon Kim
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
- Correspondence: (C.-Y.K.); (H.M.C.); Tel.: +82-10-9140-0136; Fax: +82-2-455-9012
| | - Hyung Min Chung
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Korea; (J.B.); (H.K.); (J.-H.L.); (Y.-H.J.); (S.-G.L.); (K.-R.K.); (M.-S.O.)
- Mireacellbio Co., Ltd., Seoul 04795, Korea;
- Correspondence: (C.-Y.K.); (H.M.C.); Tel.: +82-10-9140-0136; Fax: +82-2-455-9012
| |
Collapse
|
25
|
Chen C, Hua D, Shi J, Tan Z, Zhu M, Tan K, Zhang L, Huang J. Porcine Immunoglobulin Fc Fused P30/P54 Protein of African Swine Fever Virus Displaying on Surface of S. cerevisiae Elicit Strong Antibody Production in Swine. Virol Sin 2020; 36:207-219. [PMID: 32915442 DOI: 10.1007/s12250-020-00278-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 07/13/2020] [Indexed: 12/14/2022] Open
Abstract
African swine fever virus (ASFV) infects domestic pigs and European wild boars with strong, hemorrhagic and high mortality. The primary cellular targets of ASFV is the porcine macrophages. Up to now, no commercial vaccine or effective treatment available to control the disease. In this study, three recombinant Saccharomyces cerevisiae (S. cerevisiae) strains expressing fused ASFV proteins-porcine Ig heavy chains were constructed and the immunogenicity of the S. cerevisiae-vectored cocktail ASFV feeding vaccine was further evaluated. To be specific, the P30-Fcγ and P54-Fcα fusion proteins displaying on surface of S. cerevisiae cells were produced by fusing the Fc fragment of porcine immunoglobulin IgG1 or IgA1 with p30 or p54 gene of ASFV respectively. The recombinant P30-Fcγ and P54-Fcα fusion proteins expressed by S. cerevisiae were verified by Western blotting, flow cytometry and immunofluorescence assay. Porcine immunoglobulin Fc fragment fused P30/P54 proteins elicited P30/P54-specific antibody production and induced higher mucosal immunity in swine. The absorption and phagocytosis of recombinant S. cerevisiae strains in IPEC-J2 cells or porcine alveolar macrophage (PAM) cells were significantly enhanced, too. Here, we introduce a kind of cheap and safe oral S. cerevisiae-vectored vaccine, which could activate the specific mucosal immunity for controlling ASFV infection.
Collapse
Affiliation(s)
- Chen Chen
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Deping Hua
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Jingxuan Shi
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Zheng Tan
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Min Zhu
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Kun Tan
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Lilin Zhang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
26
|
Lan H, Suzuki H, Nagatake T, Hosomi K, Ikegami K, Setou M, Kunisawa J. Impaired mucociliary motility enhances antigen-specific nasal IgA immune responses to a cholera toxin-based nasal vaccine. Int Immunol 2020; 32:559-568. [PMID: 32347929 PMCID: PMC9262165 DOI: 10.1093/intimm/dxaa029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 04/24/2020] [Indexed: 11/12/2022] Open
Abstract
Nasal mucosal tissues are equipped with physical barriers, mucus and cilia, on their surface. The mucus layer captures inhaled materials, and the cilia remove the inhaled materials from the epithelial layer by asymmetrical beating. The effect of nasal physical barriers on the vaccine efficacy remains to be investigated. Tubulin tyrosine ligase-like family, member 1 (Ttll1) is an essential enzyme for appropriate movement of the cilia on respiratory epithelium, and its deficiency (Ttll1-KO) leads to mucus accumulation in the nasal cavity. Here, when mice were intra-nasally immunized with pneumococcal surface protein A (PspA, as vaccine antigen) together with cholera toxin (CT, as mucosal adjuvant), Ttll1-KO mice showed higher levels of PspA-specific IgA in the nasal wash and increased numbers of PspA-specific IgA-producing plasma cells in the nasal passages when compared with Ttll1 hetero (He) mice. Mucus removal by N-acetylcysteine did not affect the enhanced immune responses in Ttll1-KO mice versus Ttll1-He mice. Immunohistological and flow cytometry analyses revealed that retention time of PspA in the nasal cavity in Ttll1-KO mice was longer than that in Ttll1-He mice. Consistently, uptake of PspA by dendritic cells was higher in the nasopharynx-associated lymphoid tissue (NALT) of Ttll1-KO mice than that of Ttll1-He mice. These results indicate that the ciliary function of removing vaccine antigen from the NALT epithelial layer is a critical determinant of the efficacy of nasal vaccine.
Collapse
Affiliation(s)
- Huangwenxian Lan
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hidehiko Suzuki
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Koji Ikegami
- Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Mitsutoshi Setou
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, Kobe University, Hyogo, Japan
- Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
27
|
Yamamoto Y, Morozumi T, Takahashi T, Saruta J, To M, Sakaguchi W, Shimizu T, Kubota N, Tsukinoki K. Faster Short-Chain Fatty Acid Absorption from the Cecum Following Polydextrose Ingestion Increases the Salivary Immunoglobulin A Flow Rate in Rats. Nutrients 2020; 12:nu12061745. [PMID: 32545166 PMCID: PMC7353249 DOI: 10.3390/nu12061745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/25/2020] [Accepted: 06/03/2020] [Indexed: 12/20/2022] Open
Abstract
Salivary immunoglobulin A (IgA) plays a vital role in preventing upper respiratory tract infections (URTI). In our previous study, we showed that the intake of carbohydrates increases the intestinal levels of short-chain fatty acids (SCFAs), which in turn increase salivary IgA levels. However, the mechanism underlying this phenomenon has not been fully elucidated. In this study, we investigated in rats the effect of polydextrose (PDX) ingestion on salivary IgA level and SCFA concentration in cecal digesta and the portal vein. Five-week-old rats were fed with a fiber-free diet (control) or with 40 g/kg of PDX for 28 days. Compared to the control, ingestion of PDX led to a higher salivary IgA flow rate (p = 0.0013) and a higher concentration of SCFAs in the portal vein (p = 0.004). These two data were positively correlated (rs = 0.88, p = 0.0002, n = 12). In contrast, the concentration of SCFAs in cecal digesta and cecal digesta viscosity were significantly lower following PDX ingestion, compared to the control (p = 0.008 and 0.05, respectively). These findings suggest that the ingestion of PDX increases the absorption rate of SCFAs in the intestine through PDX-induced fermentation, which is accompanied by an increase in SCFA levels in the blood, and ultimately leads to increased salivary IgA levels.
Collapse
Affiliation(s)
- Yuko Yamamoto
- Department of Junior College, School of Dental Hygiene, Kanagawa Dental University, 82 Inaoka, Yokosuka 2388580, Kanagawa, Japan;
| | - Toshiya Morozumi
- Division of Periodontology, Department of Oral Interdisciplinary Medicine, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka, Yokosuka 2388580, Kanagawa, Japan;
| | - Toru Takahashi
- Department of Health and Nutrition, Faculty of Human Health, Kanazawa Gakuin University, 10 Sue-machi, Kanazawa 9201392, Ishikawa, Japan;
| | - Juri Saruta
- Division of Environmental Pathology, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka, Yokosuka 2388580, Kanagawa, Japan; (J.S.); (W.S.); (N.K.)
| | - Masahiro To
- Division of Dental Anatomy, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka, Yokosuka 2388580, Kanagawa, Japan;
| | - Wakako Sakaguchi
- Division of Environmental Pathology, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka, Yokosuka 2388580, Kanagawa, Japan; (J.S.); (W.S.); (N.K.)
| | - Tomoko Shimizu
- Department of Highly Advanced Stomatology, Graduate School of Dentistry, Kanagawa Dental University, 3-31-6 Tsuruya, Kanagawa-ku, Yokohama 2210835, Kanagawa, Japan;
| | - Nobuhisa Kubota
- Division of Environmental Pathology, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka, Yokosuka 2388580, Kanagawa, Japan; (J.S.); (W.S.); (N.K.)
| | - Keiichi Tsukinoki
- Division of Environmental Pathology, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka, Yokosuka 2388580, Kanagawa, Japan; (J.S.); (W.S.); (N.K.)
- Correspondence: ; Tel.: +81-46-822-8866
| |
Collapse
|
28
|
Li M, Wang Y, Sun Y, Cui H, Zhu SJ, Qiu HJ. Mucosal vaccines: Strategies and challenges. Immunol Lett 2019; 217:116-125. [PMID: 31669546 DOI: 10.1016/j.imlet.2019.10.013] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/08/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023]
Abstract
Mucosal immunization has potential benefits over conventional parenteral immunization, eliciting immune defense in both mucosal and systemic tissue for protecting from pathogen invasion at mucosal surfaces. To provide a first line of protection at these entry ports, mucosal vaccines have been developed and hold a significant promise for reducing the burden of infectious diseases. However, until very recently, only limited mucosal vaccines are available. This review summarizes recent advances in selected aspects regarding mucosal vaccination, including appropriate administration routes, reasonable formulations, antigen-sampling and immune responses of mucosal immunity, and the strategies used to improve mucosal vaccine efficacy. Finally, the challenges of developing successful mucosal vaccines and the potential solutions are discussed.
Collapse
Affiliation(s)
- Miao Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yi Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Yuan Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hongyu Cui
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Shu J Zhu
- College of Animal Science, Zhejiang University, Hangzhou, China.
| | - Hua-Ji Qiu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China.
| |
Collapse
|
29
|
Lan H, Hosomi K, Kunisawa J. Clostridium perfringens enterotoxin-based protein engineering for the vaccine design and delivery system. Vaccine 2019; 37:6232-6239. [PMID: 31466706 DOI: 10.1016/j.vaccine.2019.08.032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 08/02/2019] [Indexed: 02/07/2023]
Abstract
Clostridium perfringens is a major cause of food poisoning worldwide, with its enterotoxin (CPE) being the major virulence factor. The C-terminus of CPE (C-CPE) is non-toxic and is the part of the toxin that binds to epithelial cells via the claudins in tight junctions; however, C-CPE has low antigenicity. To address this issue, we have used protein engineering technology to augment the antigenicity of C-CPE and have developed a C-CPE-based vaccine against C. perfringens-mediated food poisoning. Moreover, C-CPE has properties that make it potentially useful for the development of vaccines against other bacterial toxins that cause food poisoning. For example, we hypothesized that the ability of C-CPE to bind to claudins could be harnessed to deliver vaccine antigens directly to mucosa-associated lymphoid tissues, and we successfully developed a nasally administered C-CPE-based vaccine delivery system that promotes antigen-specific mucosal and systemic immune responses. In addition, our group has revealed the roles that the nasal mucus plays in lowering the efficacy of C-CPE-based nasal vaccines. Here, we review recent advances in the development of C-CPE-based vaccines against the major bacterial toxins that cause food poisoning and discuss our C-CPE-based nasal vaccine delivery system.
Collapse
Affiliation(s)
- Huangwenxian Lan
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan; Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan; Division of Mucosal Immunology, Department of Microbiology and Immunology and International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Kobe University Graduate School of Medicine, Hyogo, Japan; Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Osaka, Japan.
| |
Collapse
|
30
|
Hashizume-Takizawa T, Kobayashi R, Tsuzukibashi O, Saito M, Kurita-Ochiai T. CCR7-deficient mice exhibit a delayed antigen-specific mucosal IgA antibody response to an oral recombinant Salmonella strain. Pathog Dis 2019; 77:5480464. [DOI: 10.1093/femspd/ftz024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 04/25/2019] [Indexed: 12/11/2022] Open
Abstract
ABSTRACT
The migration of antigen (Ag)-loading dendritic cells (DCs) from Peyer's patches (PPs) to the draining mesenteric lymph nodes (MLNs) via chemokine receptor 7 (CCR7) is thought to be an important step in the initiation of acquired immunity. Our previous study showed that PPs were indispensable for Ag-specific secretory (S)IgA antibody (Ab) responses against oral recombinant Salmonella (rSalmonella). In this study, we attempted to show direct PP DC migration to MLNs by employing photoconvertible protein transgenic mice and investigated the role of the CCR7 signaling pathway in mucosal IgA induction. Our results demonstrated an actual flux of DCs from PPs to MLNs. The frequency of CCR7+ CD11c+ DCs in MLNs of PP-deficient mice was reduced, suggesting that some PP DCs migrated via CCR7. Immunization of CCR7−/− mice elicited significantly lower levels of Ag-specific SIgA Ab responses, which was associated with diminished formation of the germinal center in PPs. However, increased SIgA Ab production and dissemination of rSalmonella were observed at later time points. These results suggest that, although CCR7 was required for SIgA induction at normal velocity, the CCR7-mediated pathway is not essential for the induction of Ag-specific SIgA Ab responses to rSalmonella.
Collapse
Affiliation(s)
- Tomomi Hashizume-Takizawa
- Departments of Microbiology and Immunology, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271-8587, Japan
| | - Ryoki Kobayashi
- Community Oral Health, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271–8587, Japan
| | - Osamu Tsuzukibashi
- Division of Laboratory Medicine for Dentistry, Department of Oral Health Science, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271–8587, Japan
| | - Masanori Saito
- Departments of Microbiology and Immunology, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271-8587, Japan
| | - Tomoko Kurita-Ochiai
- Departments of Microbiology and Immunology, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakaecho-Nishi, Matsudo, Chiba 271-8587, Japan
| |
Collapse
|
31
|
Huang J, Luo S, Huang M, Zhang T, Min Z, Liu C, Zhang Q, Yang J, Min X. Protection against fatal pneumonia through mucosal and subcutaneous immunization with the pneumococcal SP0148 protein. Microb Pathog 2019; 129:206-212. [PMID: 30772476 DOI: 10.1016/j.micpath.2019.02.018] [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: 12/03/2018] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
Streptococcus pneumoniae infection is associated with very high morbidity and mortality throughout the world. Vaccines are an effective measure for the reduction of S. pneumoniae infection. In particular, protein vaccines are attracting increasing attention because of their good immunogenicity and wide coverage of serotypes. Therefore, identifying effective protein vaccine targets is important for protein vaccine development. SP0148 is a promising protein vaccine target for S. pneumoniae and is capable of reducing S. pneumoniae colonization in the nasopharynx of mice through the IL-17A pathway. However, the protective effects of SP0148 in fatal pneumococcal infection have not been evaluated. This study used subcutaneous and nasal immunization routes to systematically evaluate the protective effects of the SP0148 protein in fatal pneumococcal infection. Subcutaneous and nasal mucosal immunization with recombinant SP0148 protein produced effective immune protection against infection with a lethal dose of S. pneumoniae and significantly prolonged survival time and increased the survival rate of mice. Furthermore, nasal immunization with SP0148 induced mouse splenocytes to secrete high levels of the cytokines IFN-γ and IL-17A. Both recombinant SP0148 protein and its antiserum inhibited the adhesion of S.pneumoniae D39 to A549 human lung epithelial cells in a dose-dependent manner. In summary, SP0148 induced mice to produce protective immune responses to fatal S. pneumoniae infection, and our results could contribute to the accumulating data on the use of SP0148 protein vaccines.
Collapse
Affiliation(s)
- Jian Huang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Shilu Luo
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Meirong Huang
- Department of Blood Transfusion, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Tao Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Zongsu Min
- Zunyi Maternal and Child Health Hospital, Zunyi, 563000, China
| | - Changjin Liu
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Qing Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Jianru Yang
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China
| | - Xun Min
- Department of Laboratory Medicine, The First Affiliated Hospital, Zunyi Medical University, Zunyi, 563003, China.
| |
Collapse
|
32
|
Kiyono H, Izuhara K. New trends in mucosal immunology and allergy. Allergol Int 2019; 68:1-3. [PMID: 30591151 DOI: 10.1016/j.alit.2018.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Indexed: 01/03/2023] Open
|
33
|
Yamamoto Y, Kubota N, Takahashi T, To M, Hayashi T, Shimizu T, Kamata Y, Saruta J, Tsukinoki K. Continuous combined intake of polydextrose and lactitol stimulates cecal fermentation and salivary IgA secretion in rats. J Oral Sci 2018; 59:603-610. [PMID: 29279570 DOI: 10.2334/josnusd.16-0820] [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] [Indexed: 11/01/2022]
Abstract
Immunoglobulin A (IgA), which plays an important role in infection defense, is upregulated in the large intestine and oral cavity through dietary fiber intake. However, the mechanism underlying salivary IgA increase through dietary fiber intake remains unknown. This study investigated time-dependent effects of non-absorbable polydextrose (PDX) and lactitol intake on salivary IgA secretion and cecal fermentation. Five-week-old rats were fed a fiber-free diet with or without 25 g/kg PDX and 25 g/kg lactitol for 1, 4, and 8 weeks. Compared to control, those who ingested PDX and lactitol had higher salivary IgA flow rates per weight of submandibular gland tissue at 4 and 8 weeks (P < 0.05), greater cecal weight and digesta at 1, 4, and 8 weeks (P < 0.05), and lower concentrations of short chain fatty acids (SCFAs) in cecal digesta (P = 0.0003). These findings suggest that the consumption of PDX and lactitol may upregulate salivary IgA secretion possibly by stimulating absorption of SCFAs produced by cecal fermentation. Thus, continuous ingestion of PDX and lactitol for up to 4 weeks could increase salivary IgA and promote immune defense against pathogen invasion through the oral route.
Collapse
Affiliation(s)
- Yuko Yamamoto
- Department of Dental Hygiene, Kanagawa Dental University, Junior College
| | - Nobuhisa Kubota
- Division of Environmental Pathology, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University
| | - Toru Takahashi
- Department of Nutrition and Health Sciences, Fukuoka Women's University
| | - Masahiro To
- Division of Dental Anatomy, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University
| | - Takashi Hayashi
- Division of Environmental Pathology, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University
| | - Tomoko Shimizu
- Department of Highly Advanced Stomatology, Graduate School of Dentistry, Kanagawa Dental University
| | - Yohei Kamata
- Department of Highly Advanced Stomatology, Graduate School of Dentistry, Kanagawa Dental University
| | - Juri Saruta
- Division of Environmental Pathology, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University
| | - Keiichi Tsukinoki
- Division of Environmental Pathology, Department of Oral Science, Graduate School of Dentistry, Kanagawa Dental University
| |
Collapse
|
34
|
Yamamoto Y, Fujino K, Saruta J, Takahashi T, To M, Fuchida S, Shimizu T, Kamata Y, Misawa K, Tsukinoki K. Effects of yogurt fermented with Lactobacillus delbrueckii
ssp. bulgaricus
OLL1073R-1 on the IgA flow rate of saliva in elderly persons residing in a nursing home: A before-after non-randomised intervention study. Gerodontology 2017; 34:479-485. [DOI: 10.1111/ger.12296] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Yuko Yamamoto
- Department of Junior College; School of Dental Hygiene; Kanagawa Dental University; Yokosuka Japan
| | - Kazuhiro Fujino
- Department of Oral Science; Division of Environmental Pathology; Graduate School of Dentistry; Kanagawa Dental UniversityGraduate School of Dentistry; Yokosuka Japan
| | - Juri Saruta
- Department of Oral Science; Division of Environmental Pathology; Graduate School of Dentistry; Kanagawa Dental UniversityGraduate School of Dentistry; Yokosuka Japan
| | - Toru Takahashi
- Department of Nutrition and Health Sciences; Fukuoka Women's University; Fukuoka UK
| | - Masahiro To
- Division of Dental Anatomy; Department of Oral Science; Graduate School of Dentistry; Kanagawa Dental University; Graduate School of Dentistry; Yokosuka Japan
| | - Shinya Fuchida
- Department of Dental Sociology; Graduate School of Dentistry; Kanagawa Dental University; Graduate School of Dentistry; Yokosuka Japan
| | - Tomoko Shimizu
- Department of Highly Advanced Stomatology; Graduate School of Dentistry; Kanagawa Dental University; Graduate School of Dentistry; Yokosuka Japan
| | - Yohei Kamata
- Department of Highly Advanced Stomatology; Graduate School of Dentistry; Kanagawa Dental University; Graduate School of Dentistry; Yokosuka Japan
| | - Kyoko Misawa
- Elderly Nursing Home; Samukawa Home; Samukawa Japan
| | - Keiichi Tsukinoki
- Department of Oral Science; Division of Environmental Pathology; Graduate School of Dentistry; Kanagawa Dental UniversityGraduate School of Dentistry; Yokosuka Japan
| |
Collapse
|
35
|
Incorporation of a bi-functional protein FimH enhances the immunoprotection of chitosan-pVP1 vaccine against coxsackievirus B3-induced myocarditis. Antiviral Res 2017; 140:121-132. [DOI: 10.1016/j.antiviral.2017.01.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 01/12/2023]
|
36
|
Aran K, Chooljian M, Paredes J, Rafi M, Lee K, Kim AY, An J, Yau JF, Chum H, Conboy I, Murthy N, Liepmann D. An oral microjet vaccination system elicits antibody production in rabbits. Sci Transl Med 2017; 9:eaaf6413. [PMID: 28275153 DOI: 10.1126/scitranslmed.aaf6413] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 08/16/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
Abstract
Noninvasive immunization technologies have the potential to revolutionize global health by providing easy-to-administer vaccines at low cost, enabling mass immunizations during pandemics. Existing technologies such as transdermal microneedles are costly, deliver drugs slowly, and cannot generate mucosal immunity, which is important for optimal immunity against pathogens. We present a needle-free microjet immunization device termed MucoJet, which is a three-dimensional microelectromechanical systems-based drug delivery technology. MucoJet is administered orally, placed adjacent to the buccal tissue within the oral cavity, and uses a self-contained gas-generating chemical reaction within its two-compartment plastic housing to produce a high-pressure liquid jet of vaccine. We show that the vaccine jet ejected from the MucoJet device is capable of penetrating the buccal mucosal layer in silico, in porcine buccal tissue ex vivo, and in rabbits in vivo. Rabbits treated with ovalbumin by MucoJet delivery have antibody titers of anti-ovalbumin immunoglobulins G and A in blood serum and buccal tissue, respectively, that are three orders of magnitude higher than rabbits receiving free ovalbumin delivered topically by a dropper in the buccal region. MucoJet has the potential to accelerate the development of noninvasive oral vaccines, given its ability to elicit antibody production that is detectable locally in the buccal tissue and systemically via the circulation.
Collapse
Affiliation(s)
- Kiana Aran
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA 91711, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Marc Chooljian
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| | - Jacobo Paredes
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
- Center of Studies and Technical Research of Gipuzkoa and Tecnun (Technological Campus of the University of Navarra), 20018 San Sebastián, Spain
| | - Mohammad Rafi
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Kunwoo Lee
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
| | - Allison Y Kim
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Jeanny An
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Jennifer F Yau
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Helen Chum
- Office of Laboratory Animal Care, UC Berkeley, Berkeley, CA 94720, USA
| | - Irina Conboy
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Niren Murthy
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| | - Dorian Liepmann
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| |
Collapse
|
37
|
Yamamoto Y, Takahahi T, To M, Nakagawa Y, Hayashi T, Shimizu T, Kamata Y, Saruta J, Tsukinoki K. The Salivary IgA Flow Rate Is Increased by High Concentrations of Short-Chain Fatty Acids in the Cecum of Rats Ingesting Fructooligosaccharides. Nutrients 2016; 8:nu8080500. [PMID: 27548207 PMCID: PMC4997413 DOI: 10.3390/nu8080500] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/21/2016] [Accepted: 08/11/2016] [Indexed: 11/22/2022] Open
Abstract
Salivary immunoglobulin A (IgA) serves as a major effector in mucosal immunity by preventing submucosal invasion of pathogens. However, the mechanism by which consumption of fermentable fibers increases IgA in saliva was not fully elucidated. This study investigated the effects of fructooligosaccharides (FOS) intake and time after feeding on IgA levels in the saliva and cecal digesta and on the concentration of short-chain fatty acids (SCFA) in the cecum in rats. Five-week-old rats were fed a fiber-free diet or a diet with 50 g/kg FOS for zero, one, four, and eight weeks. Ingestion of FOS at one and eight weeks led to a higher IgA flow rate of saliva per weight of submandibular gland tissue (p < 0.05), which positively correlated with the concentration of SCFA in the cecal digesta (rs = 0.86, p = 0.0006, n = 12), but showed no correlation with the concentration of IgA in the cecal digesta (rs = 0.15, p = 0.3, n = 48). These results suggested that ingestion of FOS increased salivary IgA secretion through high levels of SCFA in the large intestine, which was produced by fermentation of FOS. Thus, continuously ingesting FOS for more than one week could increase secretion of salivary IgA.
Collapse
Affiliation(s)
- Yuko Yamamoto
- School of Dental Hygiene, Department of Junior College, Kanagawa Dental University, Yokosuka, Kanagawa 238-8580, Japan.
| | - Toru Takahahi
- Department of Nutrition and Health Sciences, Fukuoka Women's University, Fukuoka 813-8529, Japan.
| | - Masahiro To
- Division of Environmental Pathology, Department of Oral Science, Kanagawa Dental University, Graduate School of Dentistry, Yokosuka, Kanagawa 238-8580, Japan.
| | - Yusuke Nakagawa
- Division of Environmental Pathology, Department of Oral Science, Kanagawa Dental University, Graduate School of Dentistry, Yokosuka, Kanagawa 238-8580, Japan.
| | - Takashi Hayashi
- Division of Environmental Pathology, Department of Oral Science, Kanagawa Dental University, Graduate School of Dentistry, Yokosuka, Kanagawa 238-8580, Japan.
| | - Tomoko Shimizu
- Department of Highly Advanced Stomatology, Kanagawa Dental University, Graduate School of Dentistry, Yokohama, Kanagawa 221-0835, Japan.
| | - Yohei Kamata
- Department of Highly Advanced Stomatology, Kanagawa Dental University, Graduate School of Dentistry, Yokohama, Kanagawa 221-0835, Japan.
| | - Juri Saruta
- Division of Environmental Pathology, Department of Oral Science, Kanagawa Dental University, Graduate School of Dentistry, Yokosuka, Kanagawa 238-8580, Japan.
| | - Keiichi Tsukinoki
- Division of Environmental Pathology, Department of Oral Science, Kanagawa Dental University, Graduate School of Dentistry, Yokosuka, Kanagawa 238-8580, Japan.
| |
Collapse
|