1
|
Huang S, Zhang Y, Wang T, Li X. Molecular weight-mediated interaction changes for enhancing structural stability, release behavior and M cells-targeting transport efficacy of starch-based nanoparticles. Carbohydr Polym 2024; 346:122639. [PMID: 39245530 DOI: 10.1016/j.carbpol.2024.122639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/10/2024] [Accepted: 08/18/2024] [Indexed: 09/10/2024]
Abstract
Molecular weight (Mw) of ligand-mediated nanocarriers plays a pivotal role in their architecture and properties. In this study, self-assembled ovalbumin (OVA)-loaded nanoparticles were meticulously engineered by starch polyelectrolytes with different Mw. Results unveiled that, tailoring Mw of GRGDS pentapeptides-grafted carboxymethyl starch (G-CMS) displayed strong binding-affinity and transport efficiency through microfold cells (M cells) pathway in the simulated intestinal epithelial cell monolayer in which M cells were randomly located in the Caco-2 cells monolayer. Notably, nanoparticles assembled from G-CMS with relatively higher Mw exhibited more compact structures due to the stronger interactions between layers compared to that with relatively lower Mw, which rendered remarkably stable and only 19.01 % in vitro OVA leakage under conditions of the upper gastrointestinal tract. Subsequently, more intact nanoparticles reached M cells after in vitro digestion and exhibited higher transport efficiency through the M cells pathways (apparent permeability: 9.38 × 10-5 cm/s) than Caco-2 cells, attributing to specific- and non-specific binding affinity towards M cells. Therefore, optimal Mw tailoring of starch polyelectrolytes can mediate the molecular interactions among their assembled layers and the interactions with M cells to balance the structural compactness, release and transport efficacy of nanoparticles, holding promise for advancing M cells-targeting oral delivery technologies.
Collapse
Affiliation(s)
- Shuangxia Huang
- Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yiping Zhang
- Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China; Henan Engineering Laboratory for Bioconversion Technology of Functional Microbes, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Tianxing Wang
- Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Xiaoxi Li
- Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
2
|
Khaire OT, Mhaske A, Prasad AG, Almalki WH, Srivastava N, Kesharwani P, Shukla R. State-of-the-art drug delivery system to target the lymphatics. J Drug Target 2024; 32:347-364. [PMID: 38253594 DOI: 10.1080/1061186x.2024.2309671] [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: 09/08/2023] [Accepted: 01/07/2024] [Indexed: 01/24/2024]
Abstract
PRIMARY OBJECTIVE The primary objective of the review is to assess the potential of lymphatic-targeted drug delivery systems, with a particular emphasis on their role in tumour therapy and vaccination efficacy. REASON FOR LYMPHATIC TARGETING The lymphatic system's crucial functions in maintaining bodily equilibrium, regulating metabolism, and orchestrating immune responses make it an ideal target for drug delivery. Lymph nodes, being primary sites for tumour metastasis, underscore the importance of targeting the lymphatic system for effective treatment. OUTCOME Nanotechnologies and innovative biomaterials have facilitated the development of lymphatic-targeted drug carriers, leveraging endogenous macromolecules to enhance drug delivery efficiency. Various systems such as liposomes, micelles, inorganic nanomaterials, hydrogels, and nano-capsules demonstrate significant potential for delivering drugs to the lymphatic system. CONCLUSION Understanding the physiological functions of the lymphatic system and its involvement in diseases underscores the promise of targeted drug delivery in improving treatment outcomes. The strategic targeting of the lymphatic system presents opportunities to enhance patient prognosis and advance therapeutic interventions across various medical contexts, indicating the importance of ongoing research and development in this area.
Collapse
Affiliation(s)
- Omkar T Khaire
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| | - Akshada Mhaske
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| | - Aprameya Ganesh Prasad
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| |
Collapse
|
3
|
Park JI, Cho SW, Kang JH, Park TE. Intestinal Peyer's Patches: Structure, Function, and In Vitro Modeling. Tissue Eng Regen Med 2023; 20:341-353. [PMID: 37079198 PMCID: PMC10117255 DOI: 10.1007/s13770-023-00543-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/21/2023] [Accepted: 04/06/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGOUND Considering the important role of the Peyer's patches (PPs) in gut immune balance, understanding of the detailed mechanisms that control and regulate the antigens in PPs can facilitate the development of immune therapeutic strategies against the gut inflammatory diseases. METHODS In this review, we summarize the unique structure and function of intestinal PPs and current technologies to establish in vitro intestinal PP system focusing on M cell within the follicle-associated epithelium and IgA+ B cell models for studying mucosal immune networks. Furthermore, multidisciplinary approaches to establish more physiologically relevant PP model were proposed. RESULTS PPs are surrounded by follicle-associated epithelium containing microfold (M) cells, which serve as special gateways for luminal antigen transport across the gut epithelium. The transported antigens are processed by immune cells within PPs and then, antigen-specific mucosal immune response or mucosal tolerance is initiated, depending on the response of underlying mucosal immune cells. So far, there is no high fidelity (patho)physiological model of PPs; however, there have been several efforts to recapitulate the key steps of mucosal immunity in PPs such as antigen transport through M cells and mucosal IgA responses. CONCLUSION Current in vitro PP models are not sufficient to recapitulate how mucosal immune system works in PPs. Advanced three-dimensional cell culture technologies would enable to recapitulate the function of PPs, and bridge the gap between animal models and human.
Collapse
Affiliation(s)
- Jung In Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, South Korea
| | - Seung Woo Cho
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, South Korea
| | - Joo H Kang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, South Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, South Korea.
| |
Collapse
|
4
|
Barrea L, Verde L, Auriemma RS, Vetrani C, Cataldi M, Frias-Toral E, Pugliese G, Camajani E, Savastano S, Colao A, Muscogiuri G. Probiotics and Prebiotics: Any Role in Menopause-Related Diseases? Curr Nutr Rep 2023; 12:83-97. [PMID: 36746877 PMCID: PMC9974675 DOI: 10.1007/s13668-023-00462-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW The aim of this review is to provide an overview of the menopause-related changes in microbiota and their role in the pathogenesis of menopause-related diseases. In addition, evidence on probiotic supplementation as a therapeutic strategy is discussed. RECENT FINDINGS The human microbiota is a complex community that lives in a mutualism relationship with the host. Menopause is associated with dysbiosis, and these changes in the composition of microbiota in different sites (gut, vaginal, and oral microbiota) might play a role in the pathogenesis of menopause-related diseases (i.e., osteoporosis, breast cancer, endometrial hyperplasia, periodontitis, and cardiometabolic diseases). The present review highlights the pivotal role of microbiota in postmenopausal women health, in particular it (a) may increase intestinal calcium absorption thus preventing osteoporosis, (b) is associated with reduced risk of breast cancer and type 1 endometrial hyperplasia, (c) reduces gingival inflammation and menopausal periodontitis, and (d) beneficially affects multiple cardiometabolic risk factors (i.e., obesity, inflammation, and blood glucose and lipid metabolism). However, whether oral probiotic supplementation might be used for the treatment of menopause-related dysbiosis requires further clarification.
Collapse
Affiliation(s)
- Luigi Barrea
- Dipartimento di Scienze Umanistiche, Centro Direzionale, Università Telematica Pegaso, Via Porzio, isola F2, 80143, Naples, Italy
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Ludovica Verde
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy.
- Department of Public Health, University of Naples Federico II, Naples, Italy.
| | - Renata Simona Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Claudia Vetrani
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Mauro Cataldi
- Section of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Evelyn Frias-Toral
- Universidad Católica Santiago de Guayaquil, Av. Pdte. Carlos Julio Arosemena Tola, Guayaquil, 090615, Ecuador
| | - Gabriella Pugliese
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166, Rome, Italy
| | - Silvia Savastano
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Annamaria Colao
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", University Federico II, Naples, Italy
| | - Giovanna Muscogiuri
- Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Department of Clinical Medicine and Surgery, Endocrinology Unit, University Medical School of Naples, Via Sergio Pansini 5, 80131, Naples, Italy
- Dipartimento di Medicina Clinica e Chirurgia, Unità di Endocrinologia, Diabetologia e Andrologia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
- Cattedra Unesco "Educazione alla salute e allo sviluppo sostenibile", University Federico II, Naples, Italy
| |
Collapse
|
5
|
Li J, Xia Q, Guo H, Fu Z, Liu Y, Lin S, Liu J. Decorating Bacteria with Triple Immune Nanoactivators Generates Tumor-Resident Living Immunotherapeutics. Angew Chem Int Ed Engl 2022; 61:e202202409. [PMID: 35403784 DOI: 10.1002/anie.202202409] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Indexed: 11/10/2022]
Abstract
An approach of decorating bacteria with triple immune nanoactivators is reported to develop tumor-resident living immunotherapeutics. Under cytocompatible conditions, tumor-specific antigens and checkpoint blocking antibodies are simultaneously conjugated onto bacterial surface and then polydopamine nanoparticles are formed via in situ dopamine polymerization. In addition to serving as a linker, polydopamine with its photothermal effect can repolarize tumor-associated macrophages to a pro-inflammatory phenotype. The linked antigens promote the maturation of dendritic cells and generate tumor-specific immune responses, while the anchored antibodies block immune checkpoints and activate cytotoxic T lymphocytes. Decorated bacteria show spatiotemporal tumor retention and proliferation-dependent drug release, achieving potent antitumor effects in two antigen-overexpressing tumor models. This work provides a versatile platform to prepare multimodal and long-acting therapeutics for cancer immunotherapy.
Collapse
Affiliation(s)
- Juanjuan Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.,School of Life Sciences, Hainan University, Haikou, 570228, China
| | - Qing Xia
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Haiyan Guo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenzhen Fu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yong Liu
- National Center for NanoScience & Technology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Sisi Lin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jinyao Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
6
|
Li J, Xia Q, Guo H, Fu Z, Liu Y, Lin S, Liu J. Decorating Bacteria with Triple Immune Nanoactivators Generates Tumor‐Resident Living Immunotherapeutics. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202202409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Juanjuan Li
- State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer Institute Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
- School of Life Sciences Hainan University Haikou 570228 China
| | - Qing Xia
- Department of Oncology Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Haiyan Guo
- State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer Institute Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Zhenzhen Fu
- State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer Institute Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Yong Liu
- National Center for NanoScience & Technology Chinese Academy of Sciences Beijing 100190 China
| | - Sisi Lin
- State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer Institute Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Jinyao Liu
- State Key Laboratory of Oncogenes and Related Genes Shanghai Cancer Institute Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine Institute of Molecular Medicine Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| |
Collapse
|
7
|
Xu H, Cai L, Hufnagel S, Cui Z. Intranasal vaccine: Factors to consider in research and development. Int J Pharm 2021; 609:121180. [PMID: 34637935 DOI: 10.1016/j.ijpharm.2021.121180] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
Most existing vaccines for human use are administered by needle-based injection. Administering vaccines needle-free intranasally has numerous advantages over by needle-based injection, but there are only a few intranasal vaccines that are currently approved for human use, and all of them are live attenuated influenza virus vaccines. Clearly, there are immunological as well as non-immunological challenges that prevent vaccine developers from choosing the intranasal route of administration. We reviewed current approved intranasal vaccines and pipelines and described the target of intranasal vaccines, i.e. nose and lymphoid tissues in the nasal cavity. We then analyzed factors unique to intranasal vaccines that need to be considered when researching and developing new intranasal vaccines. We concluded that while the choice of vaccine formulations, mucoadhesives, mucosal and epithelial permeation enhancers, and ligands that target M-cells are important, safe and effective intranasal mucosal vaccine adjuvants are needed to successfully develop an intranasal vaccine that is not based on live-attenuated viruses or bacteria. Moreover, more effective intranasal vaccine application devices that can efficiently target a vaccine to lymphoid tissues in the nasal cavity as well as preclinical animal models that can better predict intranasal vaccine performance in clinical trials are needed to increase the success rate of intranasal vaccines in clinical trials.
Collapse
Affiliation(s)
- Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Lucy Cai
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephanie Hufnagel
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States.
| |
Collapse
|
8
|
Alotaibi BS, Buabeid M, Ibrahim NA, Kharaba ZJ, Ijaz M, Murtaza G. Recent strategies driving oral biologic administration. Expert Rev Vaccines 2021; 20:1587-1601. [PMID: 34612121 DOI: 10.1080/14760584.2021.1990044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION High patient compliance, noninvasiveness, and self-administration are the leading features of vaccine delivery through the oral route. The implementation of swift mass vaccination campaigns in pandemic outbreaks fascinates the use of oral vaccination. This approach can elicit both mucosal and systemic immune responses to protect against infection at the surface of the mucosa. AREA COVERED As pathogen entry and spread mainly occurs through the gastrointestinal tract (GIT) mucosal surfaces, oral vaccination may protect and limit disease spread. Oral vaccines target various potential mucosal inductive sites in the GIT, such as the oral cavity, gastric area, and small intestine. Orally delivered vaccines having subunit and nucleic acid pass through various GIT-associated risks, such as the biodegradation of biologics and their reduced absorption. This article presents a summarized review of the existing technologies and prospects for oral vaccination. EXPERT OPINION The intestinal mucosa focuses on current approaches, while future strategies target new mucosal sites, i.e. oral cavity and stomach. Recent developments in biologic delivery through the oral route and their potential use in future oral vaccination are mainly considered.
Collapse
Affiliation(s)
- Badriyah Shadid Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, 346, UAE.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Nihal Abdalla Ibrahim
- Department of Clinical Sciences, Ajman University, Ajman, 346, UAE.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Zelal Jaber Kharaba
- Department of Clinical Sciences, College of Pharmacy, Al-Ain University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Munazza Ijaz
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore, 54000, Pakistan
| |
Collapse
|
9
|
Coffey JW, Gaiha GD, Traverso G. Oral Biologic Delivery: Advances Toward Oral Subunit, DNA, and mRNA Vaccines and the Potential for Mass Vaccination During Pandemics. Annu Rev Pharmacol Toxicol 2021; 61:517-540. [PMID: 32466690 PMCID: PMC8057107 DOI: 10.1146/annurev-pharmtox-030320-092348] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Oral vaccination enables pain-free and self-administrable vaccine delivery for rapid mass vaccination during pandemic outbreaks. Furthermore, it elicits systemic and mucosal immune responses. This protects against infection at mucosal surfaces, which may further enhance protection and minimize the spread of disease. The gastrointestinal (GI) tract presents a number of prospective mucosal inductive sites for vaccine targeting, including the oral cavity, stomach, and small intestine. However, currently available oral vaccines are effectively limited to live-attenuated and inactivated vaccines against enteric diseases. The GI tract poses a number of challenges,including degradative processes that digest biologics and mucosal barriers that limit their absorption. This review summarizes the approaches currently under development and future opportunities for oral vaccine delivery to established (intestinal) and relatively new (oral cavity, stomach) mucosal targets. Special consideration is given to recent advances in oral biologic delivery that offer promise as future platforms for the administration of oral vaccines.
Collapse
Affiliation(s)
- Jacob William Coffey
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunology, University of Melbourne, Victoria, 3000, Australia
| | - Gaurav Das Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139, USA
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| |
Collapse
|
10
|
Barbosa de Souza A, Vinícius Chaud M, Francine Alves T, Ferreira de Souza J, Andrade Santana MH. Hyaluronic Acid in the Intestinal Tract: Influence of Structure, Rheology, and Mucoadhesion on the Intestinal Uptake in Rats. Biomolecules 2020; 10:E1422. [PMID: 33050089 PMCID: PMC7601924 DOI: 10.3390/biom10101422] [Citation(s) in RCA: 4] [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: 09/02/2020] [Revised: 09/24/2020] [Accepted: 09/26/2020] [Indexed: 01/14/2023] Open
Abstract
Oral hyaluronic acid (HA) is a ubiquitous biopolymer that has gained attention as a treatment for local or systemic diseases. Here, we prepared and characterized structures of free HA (f-HA) with a high (>105 Da), intermediate (≤105 Da), and low (≤104 Da) average molar mass (MM); nanoparticles crosslinked with adipic dihydrazide (n-HA); and mixed formulations (mixed-HA) containing f-HA and n-HA. MM distribution determined the structure, hydrodynamic diameter, and zeta potential of the f-HAs. Crosslinking changed the physicochemical properties in n-HA. In vitro tack adhesion assays, using mucin tablets or a viable rat intestinal mucosa, showed better mucoadhesion with f-HA (intermediate MM) and mixed-HA (25% n-HA), especially in the jejunum segment. High MM f-HA presented negligible mucoadhesion. n-HA showed the deepest diffusion into the porous of the membranes. In vivo results showed that, except for high MM f-HA, there is an inverse relationship between rheological changes in the intestinal membrane macerates resulting from mucoadhesion and the effective intestinal permeability that led to blood clearance of the structures. We conclude that the n-HA formulations are promising for targeting other tissues, while formulations of f-HA (intermediate MM) and mixed-HA are better for treating dysbiosis.
Collapse
Affiliation(s)
- Alexandro Barbosa de Souza
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, P.O. Box 6066, Campinas 13083 852, SP, Brazil;
- Laboratory of Biomaterials and Nanotechnology, University of Sorocaba, Sorocaba 18300 000, SP, Brazil; (M.V.C.); (T.F.A.); (J.F.d.S.)
| | - Marco Vinícius Chaud
- Laboratory of Biomaterials and Nanotechnology, University of Sorocaba, Sorocaba 18300 000, SP, Brazil; (M.V.C.); (T.F.A.); (J.F.d.S.)
| | - Thais Francine Alves
- Laboratory of Biomaterials and Nanotechnology, University of Sorocaba, Sorocaba 18300 000, SP, Brazil; (M.V.C.); (T.F.A.); (J.F.d.S.)
| | - Juliana Ferreira de Souza
- Laboratory of Biomaterials and Nanotechnology, University of Sorocaba, Sorocaba 18300 000, SP, Brazil; (M.V.C.); (T.F.A.); (J.F.d.S.)
| | - Maria Helena Andrade Santana
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, P.O. Box 6066, Campinas 13083 852, SP, Brazil;
| |
Collapse
|
11
|
Overcoming the intestinal barrier: A look into targeting approaches for improved oral drug delivery systems. J Control Release 2020; 322:486-508. [DOI: 10.1016/j.jconrel.2020.04.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 12/17/2022]
|
12
|
Khan HS, Nair VR, Ruhl CR, Alvarez-Arguedas S, Galvan Rendiz JL, Franco LH, Huang L, Shaul PW, Kim J, Xie Y, Mitchell RB, Shiloh MU. Identification of scavenger receptor B1 as the airway microfold cell receptor for Mycobacterium tuberculosis. eLife 2020; 9:52551. [PMID: 32134383 PMCID: PMC7065847 DOI: 10.7554/elife.52551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb) can enter the body through multiple routes, including via specialized transcytotic cells called microfold cells (M cell). However, the mechanistic basis for M cell entry remains undefined. Here, we show that M cell transcytosis depends on the Mtb Type VII secretion machine and its major virulence factor EsxA. We identify scavenger receptor B1 (SR-B1) as an EsxA receptor on airway M cells. SR-B1 is required for Mtb binding to and translocation across M cells in mouse and human tissue. Together, our data demonstrate a previously undescribed role for Mtb EsxA in mucosal invasion and identify SR-B1 as the airway M cell receptor for Mtb.
Collapse
Affiliation(s)
- Haaris S Khan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Vidhya R Nair
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Cody R Ruhl
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Samuel Alvarez-Arguedas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jorge L Galvan Rendiz
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Luis H Franco
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States
| | - Linzhang Huang
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jiwoong Kim
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, United States
| | - Yang Xie
- Quantitative Biomedical Research Center, Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, United States
| | - Ron B Mitchell
- Department of Otolaryngology, University of Texas Southwestern Medical Center, Dallas, United States
| | - Michael U Shiloh
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
13
|
The intelligent delivery systems for bioactive compounds in foods: Physicochemical and physiological conditions, absorption mechanisms, obstacles and responsive strategies. Trends Food Sci Technol 2018. [DOI: 10.1016/j.tifs.2018.06.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
14
|
Namdee K, Khongkow M, Boonrungsiman S, Nittayasut N, Asavarut P, Temisak S, Saengkrit N, Puttipipatkhachorn S, Hajitou A, Ruxrungtham K, Yata T. Thermoresponsive Bacteriophage Nanocarrier as a Gene Delivery Vector Targeted to the Gastrointestinal Tract. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:33-44. [PMID: 30195771 PMCID: PMC6023791 DOI: 10.1016/j.omtn.2018.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 04/27/2018] [Accepted: 04/27/2018] [Indexed: 11/21/2022]
Abstract
The use of the gastrointestinal tract as a site for the local delivery of DNA is an exciting prospect. In order to obtain an effective vector capable of delivering a gene of interest to target cells to achieve sufficient and sustained transgene expression, with minimal toxicity, we developed a new generation of filamentous bacteriophage. This particular bacteriophage was genetically engineered to display an arginine-glycine-aspartic acid (RGD) motif (an integrin-binding peptide) on the major coat protein pVIII and carry a mammalian DNA cassette. One unanticipated observation is the thermoresponsive behavior of engineered bacteriophage. This finding has led us to simplify the isolation method to purify bacteriophage particles from cell culture supernatant by low-temperature precipitation. Our results showed that, in contrast to non-surface modified, the RGD-modified bacteriophage was successfully used to deliver a transgene to mammalian cells. Our in vitro model of the human intestinal follicle-associated epithelium also demonstrated that bacteriophage particles were stable in simulated gastrointestinal fluids and able to cross the human intestinal barrier. In addition, we confirmed an adjuvant property of the engineered bacteriophage to induce nitric oxide production by macrophages. In conclusion, our study demonstrated the possibility of using bacteriophage for gene transfer in the gastrointestinal tract.
Collapse
Affiliation(s)
- Katawut Namdee
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Mattaka Khongkow
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Suwimon Boonrungsiman
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Naiyaphat Nittayasut
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Paladd Asavarut
- Cancer Phage Therapy Group, Division of Brain Sciences, Imperial College London, London, UK
| | - Sasithon Temisak
- Bio Analysis Group, Chemical Metrology and Biometry Department, National Institute of Metrology (NIMT), Pathumthani, Thailand
| | - Nattika Saengkrit
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathumthani, Thailand
| | - Satit Puttipipatkhachorn
- Department of Manufacturing Pharmacy, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Amin Hajitou
- Cancer Phage Therapy Group, Division of Brain Sciences, Imperial College London, London, UK
| | - Kiat Ruxrungtham
- Vaccine and Cellular Immunology Laboratory, Vaccine Research Center (ChulaVRC) and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Teerapong Yata
- National Nanotechnology Centre (NANOTEC), National Science and Technology Development Agency, Pathumthani, Thailand.
| |
Collapse
|
15
|
Exploring the usefulness of the complex in vitro intestinal epithelial model Caco-2/HT29/Raji-B in nanotoxicology. Food Chem Toxicol 2018; 113:162-170. [PMID: 29421767 DOI: 10.1016/j.fct.2018.01.042] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/19/2018] [Accepted: 01/23/2018] [Indexed: 12/13/2022]
Abstract
The use of in vitro barrier models is gaining relevance as an alternative to animal studies in risk assessment, pharmacokinetic, and toxicological studies in general. These models permit an easier evaluation of the underlying mechanisms taking place at the molecular and cellular levels on the barrier site. Here, we report several methodological modifications of the three-dimensional in vitro intestinal epithelial model Caco-2/HT29/Raji-B for its successful application in the Nanotoxicology field. In addition, new insights in the study of specific molecular markers and new confocal microscope approaches have also been incorporated. Due to the multiple variables and parameters playing a part when the model's complexity is increased, we have monitored the barrier's formation and cell differentiation over time. Finally, the practical usability of the proposed model was tested by evaluating the action of the food additives titanium dioxide and silica dioxide nanoparticles (TiO2NPs and SiO2NPs). The NPs-associated effects were evaluated by confocal microscopy. We have demonstrated the essential role of the mucus layer in the decrease of cellular uptake, avoiding potential NPs-cell nuclei interactions.
Collapse
|
16
|
Liu L, Zhang W, Song Y, Wang W, Zhang Y, Wang T, Li K, Pan Q, Qi X, Gao Y, Gao L, Liu C, Zhang Y, Wang Y, He G, Wang X, Cui H. Recombinant Lactococcus lactis co-expressing OmpH of an M cell-targeting ligand and IBDV-VP2 protein provide immunological protection in chickens. Vaccine 2017; 36:729-735. [PMID: 29289381 DOI: 10.1016/j.vaccine.2017.12.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/22/2017] [Accepted: 12/11/2017] [Indexed: 12/20/2022]
Abstract
Infectious bursal disease virus (IBDV) is a highly contagious disease that results in enormous economic losses in the global poultry sector. Lactic acid bacteria are an appealing vehicle for the safe and effective delivery of heterologous protein antigens. Oral administration of the commensal bacterium Lactococcus lactis expressing recombinant fusion proteins has been used to elicit mucosal and systemic immune responses. In this study, a Lactococcus lactis NZ3900 strain co-expressing the outer membrane protein (Omp) H of the microfold (M) cell-targeting ligand and the viral capsid protein (VP)2 antigen of IBDV was genetically engineered, and its immunopotentiating capacity as an oral and injected vaccine in chickens was evaluated. Western blotting analysis demonstrated that VP2-OmpH was expressed in the cytoplasm of cells and had high immunoreactivity. An in vivo study showed that in the absence of any adjuvant, the recombinant L. lactis VP2-OmpH strain stimulated the immune response and protected against very virulent IBDV challenge in 100% and 80% of chickens immunized by injection and oral administration, respectively. Moreover, the antiviral neutralizing antibody titers induced by injection administration were higher than those induced by oral administration. Mucosal secretory IgA titers induced by oral administration were higher than those induced by injection administration. These results suggested that the recombinant L. lactis VP2-OmpH strain is a promising candidate vaccine to prevent IBDV infection.
Collapse
Affiliation(s)
- Linlin Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Wang Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Yuxin Song
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Wenqian Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yuan Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China; College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Tingting Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Kai Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Qing Pan
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Xiaole Qi
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yulong Gao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Li Gao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Changjun Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yanping Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Yongqiang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Gaoming He
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang 832003, China.
| | - Xiaomei Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| | - Hongyu Cui
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, China.
| |
Collapse
|
17
|
Løvmo SD, Speth MT, Repnik U, Koppang EO, Griffiths GW, Hildahl JP. Translocation of nanoparticles and Mycobacterium marinum across the intestinal epithelium in zebrafish and the role of the mucosal immune system. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 67:508-518. [PMID: 27343826 DOI: 10.1016/j.dci.2016.06.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/20/2016] [Accepted: 06/20/2016] [Indexed: 06/06/2023]
Abstract
Nano- and microparticles are promising carrier systems for oral delivery of drugs or vaccines, particularly in fish aquaculture. However, the mechanisms of uptake, trans-epithelial transport and immune response to nano/micrometer sized particles, or microorganisms such as bacteria are poorly understood in fish. Here, adult zebrafish were used to study the uptake of different nano- and microparticles and the pathogenic bacteria Mycobacterium marinum in the intestine, and their interactions with epithelial cells and the mucosal immune system. Fluorescent particles or bacteria were delivered directly into the adult zebrafish intestine by oral intubation and their localization was imaged in intestine, liver and spleen sections. Zebrafish do not appear to have M-cells, but both nanoparticles and bacteria were rapidly taken up in the intestine and transported to the liver and spleen. In each tissue, both bacteria and particles largely localized to leukocytes, presumably macrophages.
Collapse
Affiliation(s)
- Signe Dille Løvmo
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Martin Tobias Speth
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Urska Repnik
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Erling Olaf Koppang
- School of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| | - Gareth Wyn Griffiths
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| | - Jon Paul Hildahl
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| |
Collapse
|
18
|
Kim SH, Jang YS. The development of mucosal vaccines for both mucosal and systemic immune induction and the roles played by adjuvants. Clin Exp Vaccine Res 2017; 6:15-21. [PMID: 28168169 PMCID: PMC5292352 DOI: 10.7774/cevr.2017.6.1.15] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/21/2016] [Accepted: 12/23/2016] [Indexed: 01/22/2023] Open
Abstract
Vaccination is the most successful immunological practice that improves the quality of human life and health. Vaccine materials include antigens of pathogens and adjuvants potentiating the effectiveness of vaccination. Vaccines are categorized using various criteria, including the vaccination material used and the method of administration. Traditionally, vaccines have been injected via needles. However, given that most pathogens first infect mucosal surfaces, there is increasing interest in the establishment of protective mucosal immunity, achieved by vaccination via mucosal routes. This review summarizes recent developments in mucosal vaccines and their associated adjuvants.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology and Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea
| | - Yong-Suk Jang
- Department of Molecular Biology and Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju, Korea.; Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju, Korea
| |
Collapse
|
19
|
Yuasa H, Mantani Y, Masuda N, Nishida M, Kawano J, Yokoyama T, Hoshi N, Kitagawa H. Differential expression of Toll-like receptor-2, -4 and -9 in follicle-associated epithelium from epithelia of both follicle-associated intestinal villi and ordinary intestinal villi in rat Peyer's patches. J Vet Med Sci 2016; 78:1797-1804. [PMID: 27593683 PMCID: PMC5240757 DOI: 10.1292/jvms.16-0349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The expressions of Toll-like receptor (TLR) -2, -4 and -9 were immunohistochemically
investigated in the follicle-associated epithelium (FAE), and epithelia of the
follicle-associated intestinal villus (FAIV) and ordinary intestinal villus (IV) in rat
Peyer’s patch regions with no bacterial colonies on the mucous membranes. TLR-2 was
expressed in the striated borders of microvillous columnar epithelial cells (MV) in both
FAIV and IV except in the apices. However, TLR-2 expression in the striated borders was
weaker in the epithelium of the follicular side of FAIV (f-FAIV) than in epithelia of IV
and the anti-follicular side of FAIV. TLR-4 and -9 were not expressed in the FAIV and IV.
In the FAE, TLR-2, -4 and -9 were not expressed in the striated borders of MV, but the
roofs of some typical M-cells were immunopositive for all TLRs. Especially, no
TLR-positive MV were found at the FAE sites where M-cells appeared most frequently. In the
follicle-associated intestinal crypt (FAIC), immunopositivity for all TLRs was observed in
the striated borders of MV and the luminal substances. In conclusion, the lower levels of
TLR-2 in both FAE and the epithelium of f-FAIV probably reduce recognition of indigenous
bacteria. TLR-2, -4 and -9 appear not to participate directly in differentiation of MV
into M-cells, because TLRs were not expressed in any MV in the upstream region of M-cells
in FAE with no settlement of indigenous bacteria in the rat Peyer’s patches.
Collapse
Affiliation(s)
- Hideto Yuasa
- Laboratory of Histophysiology, Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo 657-8501, Japan
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Bleau C, Karelis AD, St-Pierre DH, Lamontagne L. Crosstalk between intestinal microbiota, adipose tissue and skeletal muscle as an early event in systemic low-grade inflammation and the development of obesity and diabetes. Diabetes Metab Res Rev 2015; 31:545-61. [PMID: 25352002 DOI: 10.1002/dmrr.2617] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 10/01/2014] [Accepted: 10/13/2014] [Indexed: 02/06/2023]
Abstract
Obesity is associated with a systemic chronic low-grade inflammation that contributes to the development of metabolic disorders such as cardiovascular diseases and type 2 diabetes. However, the etiology of this obesity-related pro-inflammatory process remains unclear. Most studies have focused on adipose tissue dysfunctions and/or insulin resistance in skeletal muscle cells as well as changes in adipokine profile and macrophage recruitment as potential sources of inflammation. However, low-grade systemic inflammation probably involves a complex network of signals interconnecting several organs. Recent evidences have suggested that disturbances in the composition of the gut microbial flora and alterations in levels of gut peptides following the ingestion of a high-fat diet may be a cause of low-grade systemic inflammation that may even precede and predispose to obesity, metabolic disorders or type 2 diabetes. This hypothesis is appealing because the gastrointestinal system is first exposed to nutrients and may thereby represent the first link in the chain of events leading to the development of obesity-associated systemic inflammation. Therefore, the present review will summarize the latest advances interconnecting intestinal mucosal bacteria-mediated inflammation, adipose tissue and skeletal muscle in a coordinated circuitry favouring the onset of a high-fat diet-related systemic low-grade inflammation preceding obesity and predisposing to metabolic disorders and/or type 2 diabetes. A particular emphasis will be given to high-fat diet-induced alterations of gut homeostasis as an early initiator event of mucosal inflammation and adverse consequences contributing to the promotion of extended systemic inflammation, especially in adipose and muscular tissues.
Collapse
MESH Headings
- Adipose Tissue, White/immunology
- Adipose Tissue, White/metabolism
- Animals
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/immunology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/microbiology
- Diet, High-Fat/adverse effects
- Enteritis/etiology
- Enteritis/immunology
- Enteritis/microbiology
- Enteritis/physiopathology
- Gastrointestinal Hormones/metabolism
- Gastrointestinal Microbiome
- Humans
- Immunity, Mucosal
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/microbiology
- Models, Biological
- Muscle, Skeletal/immunology
- Muscle, Skeletal/metabolism
- Myositis/etiology
- Myositis/immunology
- Myositis/microbiology
- Myositis/physiopathology
- Obesity/etiology
- Obesity/immunology
- Obesity/metabolism
- Obesity/microbiology
- Panniculitis/etiology
- Panniculitis/immunology
- Panniculitis/microbiology
- Panniculitis/physiopathology
- Systemic Vasculitis/etiology
- Systemic Vasculitis/immunology
- Systemic Vasculitis/microbiology
- Systemic Vasculitis/physiopathology
Collapse
Affiliation(s)
- Christian Bleau
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, Canada, H3C 3P8
| | - Antony D Karelis
- Department of Kinanthropology, Université du Québec à Montréal, Montreal, Canada, H3C 3P8
| | - David H St-Pierre
- Department of Kinanthropology, Université du Québec à Montréal, Montreal, Canada, H3C 3P8
| | - Lucie Lamontagne
- Department of Biological Sciences, Université du Québec à Montréal, Montreal, Canada, H3C 3P8
| |
Collapse
|
21
|
Schmitz S, Glanemann B, Garden OA, Brooks H, Chang YM, Werling D, Allenspach K. A prospective, randomized, blinded, placebo-controlled pilot study on the effect of Enterococcus faecium on clinical activity and intestinal gene expression in canine food-responsive chronic enteropathy. J Vet Intern Med 2015; 29:533-43. [PMID: 25776251 PMCID: PMC4895498 DOI: 10.1111/jvim.12563] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 01/07/2015] [Accepted: 01/28/2015] [Indexed: 01/27/2023] Open
Abstract
Background Canine chronic enteropathies (CE) are believed to be caused by an aberrant immune response towards the intestinal microbiome. Administration of probiotics can alleviate colitis in people. In vitro effects of the probiotic Enterococcus faecium NCIMB 10415 E1707 (EF) previously have been evaluated using canine cells (e.g., whole blood, intestinal biopsies), but data on in vivo efficacy are lacking. Hypothesis/Objectives Administration of EF to dogs with food‐responsive CE will improve clinical outcome and decrease the intestinal inflammatory profile. Animals Dogs diagnosed with CE were prospectively recruited to receive a hydrolyzed elimination diet plus either a synbiotic product containing EF or placebo for 6 weeks. Both veterinary staff and owners were blinded to the treatment. Methods Clinical severity index (CCECAI), clinicopathological data and gene expression using intestinal biopsies (TLR2/4/5/9, IL‐17A, IL‐22, IL‐23p19, RORC, IL‐2, IL‐12p35, TNFα, IL‐4, IFNy, IL‐10, TGFβ, IL‐1β, IL‐18, NLRP3, casp‐1, TFF1, TFF3 and PPARy) before and after 6 weeks of treatment were analyzed using linear mixed modeling. Results Of the 45 cases recruited, 12 finished the clinical trial. Seven received the synbiotic and 5 the placebo product. There was no difference between groups or treatments regarding clinical efficacy, histology scores or expression of any of the investigated genes. Conclusions and clinical importance Standard dietary treatment induced rapid clinical response in all cases. Because the study was underpowered, it was not possible to determine whether or not EF had an additional effect within the time period of 6 weeks.
Collapse
Affiliation(s)
- S Schmitz
- Department of Clinical Sciences and Services, Royal Veterinary College, University of London, North Mymms, Hertfordshire, UK
| | | | | | | | | | | | | |
Collapse
|
22
|
Spasova DS, Surh CD. Blowing on embers: commensal microbiota and our immune system. Front Immunol 2014; 5:318. [PMID: 25120539 PMCID: PMC4112811 DOI: 10.3389/fimmu.2014.00318] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 06/24/2014] [Indexed: 12/19/2022] Open
Abstract
Vertebrates have co-evolved with microorganisms resulting in a symbiotic relationship, which plays an important role in health and disease. Skin and mucosal surfaces are colonized with a diverse population of commensal microbiota, over 1000 species, outnumbering the host cells by 10-fold. In the past 40 years, studies have built on the idea that commensal microbiota is in constant contact with the host immune system and thus influence immune function. Recent studies, focusing on mutualism in the gut, have shown that commensal microbiota seems to play a critical role in the development and homeostasis of the host immune system. In particular, the gut microbiota appears to direct the organization and maturation of lymphoid tissues and acts both locally and systemically to regulate the recruitment, differentiation, and function of innate and adaptive immune cells. While the pace of research in the area of the mucosal–immune interface has certainly intensified over the last 10 years, we are still in the early days of this field. Illuminating the mechanisms of how gut microbes shape host immunity will enhance our understanding of the causes of immune-mediated pathologies and improve the design of next-generation vaccines. This review discusses the recent advances in this field, focusing on the close relationship between the adaptive immune system and commensal microbiota, a constant and abundant source of foreign antigens.
Collapse
Affiliation(s)
- Darina S Spasova
- Kellog School of Science and Technology Doctoral Program in Chemical and Biological Sciences and the Department of Immunology and Microbial Science, The Scripps Research Institute , La Jolla, CA , USA ; Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology , La Jolla, CA , USA
| | - Charles D Surh
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology , La Jolla, CA , USA ; Academy of Immunology and Microbiology, Institute of Basic Science , Pohang , South Korea ; Department of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology , Pohang , South Korea
| |
Collapse
|
23
|
Antigen targeting to M cells for enhancing the efficacy of mucosal vaccines. Exp Mol Med 2014; 46:e85. [PMID: 24626171 PMCID: PMC3972786 DOI: 10.1038/emm.2013.165] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 12/06/2013] [Indexed: 01/01/2023] Open
Abstract
Vaccination is one of the most successful applications of immunology and for a long time has depended on parenteral administration protocols. However, recent studies have pointed to the promise of mucosal vaccination because of its ease, economy and efficiency in inducing an immune response not only systemically, but also in the mucosal compartment where many pathogenic infections are initiated. However, successful mucosal vaccination requires the help of an adjuvant for the efficient delivery of vaccine material into the mucosa and the breaking of the tolerogenic environment, especially in oral mucosal immunization. Given that M cells are the main gateway to take up luminal antigens and initiate antigen-specific immune responses, understanding the role and characteristics of M cells is crucial for the development of successful mucosal vaccines. Especially, particular interest has been focused on the regulation of the tolerogenic mucosal microenvironment and the introduction of the luminal antigen into the lymphoid organ by exploiting the molecules of M cells. Here, we review the characteristics of M cells and the immune regulatory factors in mucosa that can be exploited for mucosal vaccine delivery and mucosal immune regulation.
Collapse
|
24
|
Targeted oral delivery of BmpB vaccine using porous PLGA microparticles coated with M cell homing peptide-coupled chitosan. Biomaterials 2014; 35:2365-73. [DOI: 10.1016/j.biomaterials.2013.11.073] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 11/23/2013] [Indexed: 01/17/2023]
|
25
|
Rochereau N, Drocourt D, Perouzel E, Pavot V, Redelinghuys P, Brown GD, Tiraby G, Roblin X, Verrier B, Genin C, Corthésy B, Paul S. Dectin-1 is essential for reverse transcytosis of glycosylated SIgA-antigen complexes by intestinal M cells. PLoS Biol 2013; 11:e1001658. [PMID: 24068891 PMCID: PMC3775721 DOI: 10.1371/journal.pbio.1001658] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 08/08/2013] [Indexed: 01/27/2023] Open
Abstract
This work reports the long-awaited identification of Dectin-1 and Siglec-5 as the M cell co-receptors that mediate the reverse transcytosis of secretory IgA molecules to mount a gut immune response. Intestinal microfold (M) cells possess a high transcytosis capacity and are able to transport a broad range of materials including particulate antigens, soluble macromolecules, and pathogens from the intestinal lumen to inductive sites of the mucosal immune system. M cells are also the primary pathway for delivery of secretory IgA (SIgA) to the gut-associated lymphoid tissue. However, although the consequences of SIgA uptake by M cells are now well known and described, the mechanisms whereby SIgA is selectively bound and taken up remain poorly understood. Here we first demonstrate that both the Cα1 region and glycosylation, more particularly sialic acid residues, are involved in M cell–mediated reverse transcytosis. Second, we found that SIgA is taken up by M cells via the Dectin-1 receptor, with the possible involvement of Siglec-5 acting as a co-receptor. Third, we establish that transcytosed SIgA is taken up by mucosal CX3CR1+ dendritic cells (DCs) via the DC-SIGN receptor. Fourth, we show that mucosal and systemic antibody responses against the HIV p24-SIgA complexes administered orally is strictly dependent on the expression of Dectin-1. Having deciphered the mechanisms leading to specific targeting of SIgA-based Ag complexes paves the way to the use of such a vehicle for mucosal vaccination against various infectious diseases. Secretory IgA (SIgA) antibodies are secreted into the gut lumen and are considered to be a first line of defense in protecting the intestinal epithelium from gut pathogens. SIgA patrol the mucus and are usually known to help immune tolerance via entrapping dietary antigens and microorganisms and other mechanisms. SIgA, in complex with its antigens, can also be taken back up by the intestinal epithelium in a process known as reverse transcytosis. SIgA can thereby promote the uptake and delivery of antigens from the intestinal lumen to the Gut-Associated Lymphoid Tissues (GALT), influencing inflammatory responses. This reverse transcytosis of SIgA is mediated by specialized epithelial M cells. Because M cells possess the ability to take up antigens and are therefore important to the local immune system, they are a key target for the specific delivery of novel mucosal vaccines against various diseases. M cell receptors that take up the SIgA-antigen complexes, which serve as mucosal vaccine vehicles, represent an important aspect of this vaccine strategy. The identification of SIgA receptor(s) on the surface of M cells has, however, remained elusive for more than a decade. In this study, we now identify Dectin-1 and Siglec-5 as the key receptors for M cell–mediated reverse transcytosis of SIgA complexes. We further find that the glycosylation modification, and particularly sialylation, of SIgA is required for its uptake by M cells. We show that, when administered orally in complex with SIgA, the HIV p24 antigen is taken up in a strictly Dectin-1-dependent manner to stimulate a mucosal and systemic antibody response. These findings are considered important for understanding gut immunity.
Collapse
MESH Headings
- Animals
- Antigen-Antibody Complex/immunology
- Antigen-Antibody Complex/metabolism
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- CHO Cells
- CX3C Chemokine Receptor 1
- Caco-2 Cells
- Cell Adhesion Molecules/metabolism
- Cell Line
- Cricetulus
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Glycosylation
- HIV Core Protein p24/immunology
- HeLa Cells
- Humans
- Immunoglobulin A/immunology
- Immunoglobulin A, Secretory/immunology
- Intestinal Mucosa/cytology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestines/cytology
- Lectins/metabolism
- Lectins, C-Type/biosynthesis
- Lectins, C-Type/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- N-Acetylneuraminic Acid/chemistry
- Receptors, Cell Surface/metabolism
- Receptors, Chemokine/metabolism
- Transcytosis/immunology
Collapse
Affiliation(s)
- Nicolas Rochereau
- GIMAP/EA3064, INSERM CIE3 Vaccinology, Université de Lyon, Saint-Etienne, France
| | | | | | - Vincent Pavot
- Institut de Biologie et Chimie des Protéines, FRE3310/CNRS, Université de Lyon, France
| | - Pierre Redelinghuys
- Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Gordon D. Brown
- Section of Infection and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | | | - Xavier Roblin
- GIMAP/EA3064, INSERM CIE3 Vaccinology, Université de Lyon, Saint-Etienne, France
| | - Bernard Verrier
- Institut de Biologie et Chimie des Protéines, FRE3310/CNRS, Université de Lyon, France
| | - Christian Genin
- GIMAP/EA3064, INSERM CIE3 Vaccinology, Université de Lyon, Saint-Etienne, France
| | - Blaise Corthésy
- R&D Laboratory of the Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Stéphane Paul
- GIMAP/EA3064, INSERM CIE3 Vaccinology, Université de Lyon, Saint-Etienne, France
- * E-mail:
| |
Collapse
|
26
|
Enhancement of serum and mucosal immune responses to a Haemophilus influenzae Type B vaccine by intranasal delivery. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2013; 20:1690-6. [PMID: 23986319 DOI: 10.1128/cvi.00215-13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Intranasal (i.n.) vaccination is potentially the most direct method for conveying upper respiratory and mucosal immunity to respiratory pathogens. However, for unclear reasons, vaccines introduced into the nasal sinuses often have lower efficacy than vaccines administered by the more frequently used parenteral routes. We examined i.n. vaccination in a mouse immune-response model with a commonly used Haemophilus influenzae type B vaccine (Hibv) composed of the polyribosylribitol phosphate (PRP) capsule antigen conjugated to tetanus toxoid. Intranasal vaccination with Hibv using a Toll-like receptor 4 (TLR4) agonist as an adjuvant significantly increased the levels of IgA specific for the PRP capsule antigen in blood serum, saliva, and mucosal secretion specimens. In contrast, control mice vaccinated transdermally (t.d.) with Hibv did not produce significant levels of PRP-specific IgA in the blood serum and saliva, and anti-PRP IgG was increased only in serum. The i.n. and t.d. vaccinations resulted in equivalent bactericidal antibody responses in blood serum, suggesting that vaccine-derived IgG is protective against infection. Elevated levels of IgG specific for the tetanus toxoid carrier protein were measured in nasal sinuses and vaginal secretions in mice vaccinated by either the t.d. or i.n. route. Tissue culture studies confirmed that the nasopharynx-associated lymphoid tissue (NALT) was at least one of the sources of PRP-specific IgA and carrier-specific IgG within the nasal sinuses. We conclude that i.n. vaccination aided by a TLR4 agonist results in robust immune responses to both the carrier protein and bacterial polysaccharide components of the Hibv.
Collapse
|
27
|
Kim SH, Jung DI, Yang IY, Jang SH, Kim J, Truong TT, Pham TV, Truong NU, Lee KY, Jang YS. Application of an M-cell-targeting ligand for oral vaccination induces efficient systemic and mucosal immune responses against a viral antigen. Int Immunol 2013; 25:623-32. [DOI: 10.1093/intimm/dxt029] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
28
|
Kim SH, Yang IY, Jang SH, Kim J, Truong TT, Van Pham T, Truong NU, Lee KY, Jang YS. C5a receptor-targeting ligand-mediated delivery of dengue virus antigen to M cells evokes antigen-specific systemic and mucosal immune responses in oral immunization. Microbes Infect 2013; 15:895-902. [PMID: 23892099 DOI: 10.1016/j.micinf.2013.07.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 04/03/2013] [Accepted: 07/16/2013] [Indexed: 12/16/2022]
Abstract
Oral mucosal immunization is a feasible and economic vaccination strategy. In order to achieve a successful oral mucosal vaccination, antigen delivery to gut immune inductive site and avoidance of oral tolerance induction should be secured. One promising approach is exploring the specific molecules expressed on the apical surfaces of M cells that have potential for antigen uptake and immune stimulation. We previously identified complement 5a receptor (C5aR) expression on human M-like cells and mouse M cells and confirmed its non-redundant role as a target receptor for antigen delivery to M cells using a model antigen. Here, we applied the OmpH ligand, which is capable of targeting the ligand-conjugated antigen to M cells to induce specific mucosal and systemic immunities against the EDIII of dengue virus (DENV). Oral immunization with the EDIII-OmpH efficiently targeted the EDIII to M cells and induced EDIII-specific immune responses comparable to those induced by co-administration of EDIII with cholera toxin (CT). Also, the enhanced responses by OmpH were characterized as Th2-skewed responses. Moreover, oral immunization using EDIII-OmpH did not induce systemic tolerance against EDIII. Collectively, we suggest that OmpH-mediated targeting of antigens to M cells could be used for an efficient oral vaccination against DENV infection.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology, Chonbuk National University, Jeonju 561-756, Republic of Korea; Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Asai T, Morrison SL. The SRC family tyrosine kinase HCK and the ETS family transcription factors SPIB and EHF regulate transcytosis across a human follicle-associated epithelium model. J Biol Chem 2013; 288:10395-405. [PMID: 23439650 DOI: 10.1074/jbc.m112.437475] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
A critical step in the induction of adaptive mucosal immunity is antigen transcytosis, in which luminal antigens are transported to organized lymphoid tissues across the follicle-associated epithelium (FAE) of Peyer's patches. However, virtually nothing is known about intracellular signaling proteins and transcription factors that regulate apical-to-basolateral transcytosis. The FAE can transcytose a variety of luminal contents, including inert particles, in the absence of specific opsonins. Furthermore, it expresses receptors for secretory immunoglobulin A (SIgA), the main antibody in mucosal secretions, and uses them to efficiently transcytose SIgA-opsonized particles present in the lumen. Using a human FAE model, we show that the tyrosine kinase HCK regulates apical-to-basolateral transcytosis of non-opsonized and SIgA-opsonized particles. We also show that, in cultured intestinal epithelial cells, ectopic expression of the transcription factor SPIB or EHF is sufficient to activate HCK-dependent apical-to-basolateral transcytosis of these particles. Our results provide the first molecular insights into the intracellular regulation of antigen sampling at mucosal surfaces.
Collapse
Affiliation(s)
- Tsuneaki Asai
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, California 90095, USA.
| | | |
Collapse
|
30
|
Expression patterns of porcine Toll-like receptors family set of genes (TLR1-10) in gut-associated lymphoid tissues alter with age. Res Vet Sci 2013; 95:92-102. [PMID: 23433683 DOI: 10.1016/j.rvsc.2013.01.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 01/17/2013] [Accepted: 01/25/2013] [Indexed: 01/09/2023]
Abstract
The aim was to study the expression pattern of the porcine TLR family (TLR1-10) genes in gut-associated lymphoid tissues (GALT) of varying ages. A total of nine clinically healthy pigs of three ages group (1 day, 2 months and 5 months old) were selected for this experiment (three pigs in each group). Tissues from intestinal mucosa in stomach, duodenum, jejunum and ileum and mesenteric lymph node (MLN) were used. mRNA expression of TLRs (1-10) was detectable in all tissues and TLR3 showed the highest mRNA abundance among TLRs. TLR3 expression in stomach, and TLR1 and TLR6 expression in MLN were higher in adult than newborn pigs. The western blot results of TLR2, 3 and 9 in some cases, did not coincide with the mRNA expression results. The protein localization of TLR2, 3 and 9 showed that TLR expressing cells were abundant in the lamina propria, Peyer's patches in intestine, and around and within the lymphoid follicles in the MLN. This expressions study sheds the first light on the expression patterns of all TLR genes in GALT at different ages of pigs.
Collapse
|
31
|
Moyle PM, Toth I. Modern subunit vaccines: development, components, and research opportunities. ChemMedChem 2013; 8:360-76. [PMID: 23316023 DOI: 10.1002/cmdc.201200487] [Citation(s) in RCA: 331] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 12/08/2012] [Indexed: 12/11/2022]
Abstract
Traditional vaccines, based on the administration of killed or attenuated microorganisms, have proven to be among the most effective methods for disease prevention. Safety issues related to administering these complex mixtures, however, prevent their universal application. Through identification of the microbial components responsible for protective immunity, vaccine formulations can be simplified, enabling molecular-level vaccine characterization, improved safety profiles, prospects to develop new high-priority vaccines (e.g. for HIV, tuberculosis, and malaria), and the opportunity for extensive vaccine component optimization. This subunit approach, however, comes at the expense of decreased immunity, requiring the addition of immunostimulatory agents (adjuvants). As few adjuvants are currently used in licensed vaccines, adjuvant development represents an exciting area for medicinal chemists to play a role in the future of vaccine development. In addition, immune responses can be further customized though optimization of delivery systems, tuning the size of particulate vaccines, targeting specific cells of the immune system (e.g. dendritic cells), and adding components to aid vaccine efficacy in whole immunized populations (e.g. promiscuous T-helper epitopes). Herein we review the current state of the art and future direction in subunit vaccine development, with a focus on the described components and their potential to steer the immune response toward a desired response.
Collapse
Affiliation(s)
- Peter Michael Moyle
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, QLD 4072, Australia.
| | | |
Collapse
|
32
|
Kim SH, Lee KY, Jang YS. Mucosal Immune System and M Cell-targeting Strategies for Oral Mucosal Vaccination. Immune Netw 2012; 12:165-75. [PMID: 23213309 PMCID: PMC3509160 DOI: 10.4110/in.2012.12.5.165] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 10/05/2012] [Accepted: 10/09/2012] [Indexed: 02/08/2023] Open
Abstract
Vaccination is one of the most effective methods available to prevent infectious diseases. Mucosa, which are exposed to heavy loads of commensal and pathogenic microorganisms, are one of the first areas where infections are established, and therefore have frontline status in immunity, making mucosa ideal sites for vaccine application. Moreover, vaccination through the mucosal immune system could induce effective systemic immune responses together with mucosal immunity in contrast to parenteral vaccination, which is a poor inducer of effective immunity at mucosal surfaces. Among mucosal vaccines, oral mucosal vaccines have the advantages of ease and low cost of vaccine administration. The oral mucosal immune system, however, is generally recognized as poorly immunogenic due to the frequent induction of tolerance against orally-introduced antigens. Consequently, a prerequisite for successful mucosal vaccination is that the orally introduced antigen should be transported across the mucosal surface into the mucosa-associated lymphoid tissue (MALT). In particular, M cells are responsible for antigen uptake into MALT, and the rapid and effective transcytotic activity of M cells makes them an attractive target for mucosal vaccine delivery, although simple transport of the antigen into M cells does not guarantee the induction of specific immune responses. Consequently, development of mucosal vaccine adjuvants based on an understanding of the biology of M cells has attracted much research interest. Here, we review the characteristics of the oral mucosal immune system and delineate strategies to design effective oral mucosal vaccines with an emphasis on mucosal vaccine adjuvants.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Department of Molecular Biology and the Institute for Molecular Biology and Genetics, Chonbuk National University, Jeonju 561-756, Korea
| | | | | |
Collapse
|
33
|
Sotolongo J, Ruiz J, Fukata M. The role of innate immunity in the host defense against intestinal bacterial pathogens. Curr Infect Dis Rep 2012; 14:15-23. [PMID: 22139594 DOI: 10.1007/s11908-011-0234-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Eradication of infectious disease is our global health challenge. After encountering intestinal infection with a bacterial pathogen, the host defense program is initiated by local antigen-presenting cells (APCs) that eliminate invading pathogens by phagocytosis and establish localized inflammation by secreting cytokines and chemokines. These pathogen-experienced APCs migrate to the mesenteric lymph nodes, where host immune responses are precisely orchestrated. Initiation and regulation of this defense program appear to be largely dependent on innate immunity which is antigen non-specific and provides a rapid defense against broader targets. On the other hand, many bacterial enteropathogens have evoked abilities to modify the host defense program to their advantage. Therefore, better understanding of the host-pathogen interactions is essential to establish effective eradication strategies for enteric infectious diseases. In this review, we will discuss the current understanding of innate immune regulation of the host defense mechanisms against intestinal infection by bacterial pathogens.
Collapse
Affiliation(s)
- John Sotolongo
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Post Office Box 016960 (D-149), Miami, FL, 33101, USA
| | | | | |
Collapse
|
34
|
Cisney ED, Fernandez S, Hall SI, Krietz GA, Ulrich RG. Examining the role of nasopharyngeal-associated lymphoreticular tissue (NALT) in mouse responses to vaccines. J Vis Exp 2012:3960. [PMID: 22871688 PMCID: PMC3476754 DOI: 10.3791/3960] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The nasopharyngeal-associated lymphoreticular tissues (NALT) found in humans, rodents, and other mammals, contribute to immunity in the nasal sinuses1-3. The NALT are two parallel bell-shaped structures located in the nasal passages above the hard palate, and are usually considered to be secondary components of the mucosal-associated lymphoid system4-6. Located within the NALT are discrete compartments of B and T lymphocytes interspersed with antigen-presenting dendritic cells4,7,8. These cells are surrounded by an epithelial cell layer intercalated with M-cells that are responsible for antigen retrieval from the mucosal surfaces of the air passages9,10. Naive lymphocytes circulating through the NALT are poised to respond to first encounters with respiratory pathogens7. While NALT disappear in humans by the age of two years, the Waldeyer's Ring and similarly structured lymphatic organs continue to persist throughout life6. In contrast to humans, mice retain NALT throughout life, thus providing a convenient animal model for the study of immune responses originating within the nasal sinuses11. Cultures of single-cell suspensions of NALT are not practical due to low yields of mononuclear cells. However, NALT biology can be examined by ex vivo culturing of the intact organ, and this method has the additional advantage of maintaining the natural tissue structure. For in vivo studies, genetic knockout models presenting defects limited to NALT are not currently available due to a poor understanding of the developmental pathway. For example, while lymphotoxin-α knockout mice have atrophied NALT, the Peyer's patches, peripheral lymph nodes, follicular dendritic cells and other lymphoid tissues are also altered in these genetically manipulated mice12,13. As an alternative to gene knockout mice, surgical ablation permanently eliminates NALT from the nasal passage without affecting other tissues. The resulting mouse model has been used to establish relationships between NALT and immune responses to vaccines1,3. Serial collection of serum, saliva, nasal washes and vaginal secretions is necessary for establishing the basis of host responses to vaccination, while immune responses originating directly from NALT can be confirmed by tissue culture. The following procedures outline the surgeries, tissue culture and sample collection necessary to examine local and systemic humoral immune responses to intranasal (IN) vaccination.
Collapse
Affiliation(s)
- Emily D Cisney
- U.S. Army Medical Research Institute of Infectious Diseases, USA
| | | | | | | | | |
Collapse
|
35
|
Crossing the barrier: Targeting epithelial receptors for enhanced oral vaccine delivery. J Control Release 2012; 160:431-9. [DOI: 10.1016/j.jconrel.2012.02.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 02/02/2012] [Indexed: 01/09/2023]
|
36
|
Epithelial cell coculture models for studying infectious diseases: benefits and limitations. J Biomed Biotechnol 2011; 2011:852419. [PMID: 22007147 PMCID: PMC3189631 DOI: 10.1155/2011/852419] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 07/19/2011] [Accepted: 07/27/2011] [Indexed: 12/20/2022] Open
Abstract
Countless in vitro cell culture models based on the use of epithelial cell types of single lineages have been characterized and have provided insight into the mechanisms of infection for various microbial pathogens. Diverse culture models based on disease-relevant mucosal epithelial cell types derived from gastrointestinal, genitourinary, and pulmonary organ systems have delineated many key host-pathogen interactions that underlie viral, parasitic, and bacterial disease pathogenesis. An alternative to single lineage epithelial cell monoculture, which offers more flexibility and can overcome some of the limitations of epithelial cell culture models based on only single cell types, is coculture of epithelial cells with other host cell types. Various coculture models have been described, which incorporate epithelial cell types in culture combination with a wide range of other cell types including neutrophils, eosinophils, monocytes, and lymphocytes. This paper will summarize current models of epithelial cell coculture and will discuss the benefits and limitations of epithelial cell coculture for studying host-pathogen dynamics in infectious diseases.
Collapse
|
37
|
Tyrer PC, Bean EG, Ruth Foxwell A, Pavli P. Effects of bacterial products on enterocyte-macrophage interactions in vitro. Biochem Biophys Res Commun 2011; 413:336-41. [PMID: 21893047 DOI: 10.1016/j.bbrc.2011.08.100] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 08/22/2011] [Indexed: 11/25/2022]
Abstract
We describe a coculture model of a human intestinal epithelial cell line and human peripheral blood monocytes in which monocytes differentiate into cells with features of resident intestinal macrophages. Caco-2 cells are grown on the lower surface of a semipermeable filter with pore size of 3 μm (Transwells) until they differentiate into enterocytes. Peripheral-blood monocytes are added and the co-culture incubated for two days. Monocytes migrate through the pores of the membrane, come into direct contact with the basolateral surfaces of the epithelial cell monolayer, and develop characteristics of resident intestinal macrophages including downregulation of CD14 expression and reduced pro-inflammatory cytokine responses (IL-8, TNF and IL-1β) to bacterial products. The apical application of lipopolysaccharide (LPS) and muramyl dipeptide (MDP) resulted in an increased number of integrated monocytes, but abrogated the downregulation of CD14 expression and the diminished cytokine responses. MDP also reduced tight-junctional integrity, whilst LPS had no effect. These data indicate that LPS and MDP have significant pathophysiological effects on enterocyte-monocyte interactions, and confirm other studies that demonstrate that enterocytes and their products influence monocyte differentiation. This model may be useful in providing insights into the interaction between monocytes, epithelial cells and intestinal bacteria in health and disease.
Collapse
Affiliation(s)
- Peter C Tyrer
- Inflammatory Bowel Disease Research Group, Level 5, Building 10, The Canberra Hospital, Yamba Drive, Garran, ACT 2606, Australia.
| | | | | | | |
Collapse
|
38
|
Tiwari S, Verma SK, Agrawal GP, Vyas SP. Viral protein complexed liposomes for intranasal delivery of hepatitis B surface antigen. Int J Pharm 2011; 413:211-9. [DOI: 10.1016/j.ijpharm.2011.04.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 04/09/2011] [Accepted: 04/14/2011] [Indexed: 10/18/2022]
|
39
|
Hallstrom K, McCormick BA. Salmonella Interaction with and Passage through the Intestinal Mucosa: Through the Lens of the Organism. Front Microbiol 2011; 2:88. [PMID: 21747800 PMCID: PMC3128981 DOI: 10.3389/fmicb.2011.00088] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Accepted: 04/13/2011] [Indexed: 01/13/2023] Open
Abstract
Salmonella enterica serotypes are invasive enteric pathogens spread through fecal contamination of food and water sources, and represent a constant public health threat around the world. The symptoms associated with salmonellosis and typhoid disease are largely due to the host response to invading Salmonella, and to the mechanisms these bacteria employ to survive in the presence of, and invade through the intestinal mucosal epithelia. Surmounting this barrier is required for survival within the host, as well as for further dissemination throughout the body, and subsequent systemic disease. In this review, we highlight some of the major hurdles Salmonella must overcome upon encountering the intestinal mucosal epithelial barrier, and examine how these bacteria surmount and exploit host defense mechanisms.
Collapse
Affiliation(s)
- Kelly Hallstrom
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School Worcester, MA, USA
| | | |
Collapse
|
40
|
Nasal immunity to staphylococcal toxic shock is controlled by the nasopharynx-associated lymphoid tissue. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:667-75. [PMID: 21325486 DOI: 10.1128/cvi.00477-10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The nasopharynx-associated lymphoid tissue (NALT) of humans and other mammals is associated with immunity against airborne infections, though it is generally considered to be a secondary component of the mucosa-associated lymphoid system. We found that protective immunity to a virulence factor of nasal mucosa-colonizing Staphylococcus aureus, staphylococcal enterotoxin B (SEB), requires a functional NALT. We examined the role of NALT using intranasal (IN) vaccination with a recombinant SEB vaccine (rSEBv) combined with an adjuvant in a mouse model of SEB-induced toxic shock. The rSEBv was rapidly internalized by NALT cells at the mucosal barrier, and transport into NALT was accelerated by inclusion of a Toll-like receptor 4 (TLR4) agonist. Vaccine-induced germinal centers of B cells formed within NALT, accompanied by elevated levels of IgA(+) and IgG(+) cells, and these were further increased by TLR4 activation. The NALT was the site of specific anti-rSEBv IgA and IgG production but was also influenced by intraperitoneal (IP) inoculation and perhaps other isolated lymphoid follicles observed within the nasal cavity. Vaccination by the IN route generated robust levels of anti-rSEBv IgA in saliva, nasal secretions, and blood compared to much lower levels after IP vaccination. IN vaccination also induced secretion of anti-rSEBv IgG in the blood and nasal secretions. Significantly, the efficacy of IN vaccination was dependent on NALT, as surgical removal resulted in greater sensitivity to IN challenge with wild-type SEB. Thus, protective immunity to SEB within the nasal sinuses was elicited by responses originating in NALT.
Collapse
|
41
|
Tiwari S, Agrawal GP, Vyas SP. Molecular basis of the mucosal immune system: from fundamental concepts to advances in liposome-based vaccines. Nanomedicine (Lond) 2010; 5:1617-40. [DOI: 10.2217/nnm.10.128] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The mucosal immune system, the primary portal for entry of most prevalent and devastating pathogens, is guarded by the special lymphoid tissues (mucosally associated lymphoid tissues) for immunity. Mucosal immune infection results in induction of IgA-manifested humoral immunity. Cell-mediated immunity may also be generated, marked by the presence of CD4+ Th1 and CD8+ cells. Furthermore, the immunity generated at the mucosal site is transported to the distal mucosal site as well as to systemic tissues. An understanding of the molecular basis of the mucosal immune system provides a unique platform for designing a mucosal vaccine. Coadministration of immunostimulatory molecules further accelerates functioning of the immune system. Mimicking receptor-mediated binding of the pathogen may be achieved by direct conjugation of antigen with an immunostimulatory molecule or encapsulation in a carrier followed by anchoring of a ligand having affinity to the cells of the mucosal immune system. Nanotechnology has played a significant role in mucosal vaccine development and among the available options liposomes are the most promising. Liposomes are phospholipid bilayered vesicles that can encapsulate protein as well as DNA-based vaccines and offer coencapsulation of adjuvant along with the antigen. At the same, time ligand-conjugated liposomes augment interaction of antigen with the cells of the mucosal immune system and thereby serve as suitable candidates for the mucosal delivery of vaccines. This article exhaustively explores strategies involved in the generation of mucosal immunity and also provides an insight to the progress that has been made in the development of liposome-based mucosal vaccine.
Collapse
Affiliation(s)
- Shailja Tiwari
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India
| | - Govind P Agrawal
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India
| | | |
Collapse
|
42
|
Kim SH, Seo KW, Kim J, Lee KY, Jang YS. The M cell-targeting ligand promotes antigen delivery and induces antigen-specific immune responses in mucosal vaccination. THE JOURNAL OF IMMUNOLOGY 2010; 185:5787-95. [PMID: 20952686 DOI: 10.4049/jimmunol.0903184] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Oral mucosal immunization can induce protective immunity in both systemic compartments and the mucosa. Successful mucosal immunization depends on Ag delivery to the mucosal immune induction site. The high transcytotic activity of M cells within the mucosa makes these cells attractive targets for mucosal Ag delivery, although it remains unclear whether delivery of Ag to M cells only can guarantee the induction of effective immune responses. In this study, we evaluated the ability of an M cell-targeting ligand with adjuvant activity to induce immunity against ligand-fused Ag. We selected M cell-targeting ligands through biopanning of a phage display library against differentiated in vitro M-like cells and produced the recombinant Ags fused to the selected ligands using the model Ag. One of the selected peptide ligands, Co1, promoted the binding of ligand-fused Ag to mouse Peyer's patch M cells and human M-like cells that had been defined by binding with the M cell-specific and anti-GP2 Abs. In addition, Co1 ligand enhanced the uptake of fused Ag by immunogenic tissue in an ex vivo loop assay and in vivo oral administration experiments. After oral administration, the ligand-fused Ag enhanced immune responses against the fused Ag compared with those of the control Ag without ligand. In addition, this use of the ligand supported a skewed Th2-type immune response against the fused Ag. Collectively, these results suggest that the ligand selected through biopanning against cultured M-like cells could be used as an adjuvant for targeted Ag delivery into the mucosal immune system to enhance immune induction.
Collapse
Affiliation(s)
- Sae-Hae Kim
- Division of Biological Sciences, Chonbuk National University, Jeonju, South Korea
| | | | | | | | | |
Collapse
|
43
|
|
44
|
Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function. Nat Rev Immunol 2010; 10:131-44. [PMID: 20098461 DOI: 10.1038/nri2707] [Citation(s) in RCA: 886] [Impact Index Per Article: 59.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A single layer of epithelial cells lines the small and large intestines and functions as a barrier between commensal bacteria and the rest of the body. Ligation of Toll-like receptors (TLRs) on intestinal epithelial cells by bacterial products promotes epithelial cell proliferation, secretion of IgA into the gut lumen and expression of antimicrobial peptides. As described in this Review, this establishes a microorganism-induced programme of epithelial cell homeostasis and repair in the intestine. Dysregulation of this process can result in chronic inflammatory and over-exuberant repair responses, and it is associated with the development of colon cancer. Thus, dysregulated TLR signalling by intestinal epithelial cells may explain how colonic bacteria and inflammation promote colorectal cancer.
Collapse
|
45
|
Kaminski RW, Oaks EV. Inactivated and subunit vaccines to prevent shigellosis. Expert Rev Vaccines 2010; 8:1693-704. [PMID: 19943764 DOI: 10.1586/erv.09.127] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Shigellosis remains a formidable disease globally, with children of the developing world bearing the greatest number of infections. The need for an affordable, safe and efficacious vaccine has persisted for decades. Vaccines to prevent shigellosis can be divided into living and nonliving approaches. Several nonliving Shigella vaccines are currently at different stages of development and show substantial promise. Outlined here is an overview of multiple nonliving vaccine technologies, highlighting their current status and recent advances in testing. In addition, gaps in the knowledge base regarding immune mechanisms of protection are explored.
Collapse
Affiliation(s)
- Robert W Kaminski
- Division of Bacterial and Rickettsial Diseases, Walter Reed Army Institute of Research, 503 Robert Grant Avenue, Silver Spring, MD 20910, USA
| | | |
Collapse
|
46
|
Abstract
The human intestine is colonized by an estimated 100 trillion bacteria. Some of these bacteria are essential for normal physiology, whereas others have been implicated in the pathogenesis of multiple inflammatory diseases including IBD and asthma. This review examines the influence of signals from intestinal bacteria on the homeostasis of the mammalian immune system in the context of health and disease. We review the bacterial composition of the mammalian intestine, known bacterial-derived immunoregulatory molecules, and the mammalian innate immune receptors that recognize them. We discuss the influence of bacterial-derived signals on immune cell function and the mechanisms by which these signals modulate the development and progression of inflammatory disease. We conclude with an examination of successes and future challenges in using bacterial communities or their products in the prevention or treatment of human disease.
Collapse
Affiliation(s)
- David A Hill
- University of Pennsylvania School of Veterinary Medicine, Department of Pathobiology, Philadelphia, 19104-4539, USA
| | | |
Collapse
|
47
|
Keely S, Glover LE, Weissmueller T, MacManus CF, Fillon S, Fennimore B, Colgan SP. Hypoxia-inducible factor-dependent regulation of platelet-activating factor receptor as a route for gram-positive bacterial translocation across epithelia. Mol Biol Cell 2009; 21:538-46. [PMID: 20032301 PMCID: PMC2820419 DOI: 10.1091/mbc.e09-07-0573] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Results from these studies reveal that some strains of Gram-positive bacteria exploit hypoxia-inducible factor-regulated platelet-activating factor receptor as a means for translocation through intestinal epithelial cells. Mucosal surfaces, such as the lung and intestine, are lined by a monolayer of epithelia that provides tissue barrier and transport function. It is recently appreciated that a common feature of inflammatory processes within the mucosa is hypoxia (so-called inflammatory hypoxia). Given the strong association between bacterial translocation and mucosal inflammatory disease, we hypothesized that intestinal epithelial hypoxia influences bacterial translocation. Initial studies revealed that exposure of cultured intestinal epithelia to hypoxia (pO2, 20 torr; 24–48 h) resulted in a increase of up to 40-fold in the translocation of some strains of Gram-positive bacteria, independently of epithelial barrier function. A screen of relevant pathway inhibitors identified a prominent role for the platelet-activating factor receptor (PAFr) in hypoxia-associated bacterial translocation, wherein pharmacologic antagonists of PAFr blocked bacterial translocation by as much as 80 ± 6%. Extensions of these studies revealed that hypoxia prominently induces PAFr through a hypoxia-inducible factor (HIF)-dependent mechanism. Indeed, HIF and PAFr loss of function studies (short hairpin RNA) revealed that apically expressed PAFr is central to the induction of translocation for the Gram-positive bacteria Enterococcus faecalis. Together, these findings reveal that some strains of Gram-positive bacteria exploit HIF-regulated PAFr as a means for translocation through intestinal epithelial cells.
Collapse
Affiliation(s)
- Simon Keely
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Liu CY, Mueller MH, Rogler G, Grundy D, Kreis ME. Differential afferent sensitivity to mucosal lipopolysaccharide from Salmonella typhimurium and Escherichia coli in the rat jejunum. Neurogastroenterol Motil 2009; 21:1335-e129. [PMID: 19614870 DOI: 10.1111/j.1365-2982.2009.01358.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Postinfectious irritable bowel syndrome may develop subsequent to acute bacterial enteritis. We therefore hypothesized that intestinal afferents may develop hypersensitivity upon exposure to luminal lipopolysaccharide (LPS) from pathogens but not from commensal bacteria and that this may be prostaglandin mediated. Extracellular recordings of jejunal afferents were obtained in vivo from male Wistar rats (n = 5 per group; 300-400 g). Lipopolysaccharide from Escherichia coli (E-LPS), Salmonella typhimurium (S-LPS) or vehicle were infused into the intestinal lumen at 5 mg mL(-1). The selective 5-HT(3)-receptor agonist 2-methyl-5-HT (2m5-HT, 15 microgkg(-1), i.v.) was administered at 15-min intervals before and up to 2 h after S-LPS administration. Intraluminal E-LPS had no effect on mesenteric afferent nerve discharge at baseline. By contrast, afferent discharge increased from 21.7 +/- 0.3 impsec(-1) to 28.8 +/- 3.4 impsec(-1) 40 min after S-LPS administration (mean +/- SEM; P < 0.05) and reached 38.8 +/- 4.1 impsec(-1) after 2 h (P < 0.05). The afferent response to 2m5-HT was enhanced 30 min following S-LPS by 30.9 +/- 3.9% (P < 0.05) and remained elevated thereafter. The increase in baseline discharge and sensitivity to 2m5-HT following S-LPS was prevented by pretreatment with naproxen (COX inhibitor, 10 mgkg(-1) i.v.) or AH-6809 (EP1/EP2 receptor antagonist, 1 mg kg(-1)). Intestinal afferents do not alter their discharge rate to LPS from E. coli but to LPS from the pathogenic bacterium S. typhimurium. The latter response entails afferent sensitisation to 2m5-HT that depends on prostanoid release. This acute sensitisation may prime the intestinal afferent innervation for a later development of persistent hypersensitivity.
Collapse
Affiliation(s)
- C Y Liu
- Shandong University, Department of Physiology and Key Lab of Medical Neurobiology, School of Medicine, Shandong, China
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Roles of capsule and lipopolysaccharide O antigen in interactions of human monocyte-derived dendritic cells and Klebsiella pneumoniae. Infect Immun 2009; 78:210-9. [PMID: 19841082 DOI: 10.1128/iai.00864-09] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In humans, Klebsiella pneumoniae is a saprophytic bacterium of the nasopharyngeal and intestinal mucosae that is also frequently responsible for severe nosocomial infections. Two major factors of virulence, capsular polysaccharide (CPS) and lipopolysaccharide (LPS) O antigen, are involved in mucosal colonization and the development of infections. These bacterial surface structures are likely to play major roles in interactions with the mucosal immune system, which are orchestrated by a network of surveillance based on dendritic cells (DCs). To determine the roles of K. pneumoniae CPS and LPS in the DC response, we investigated the response of immature human monocyte-derived DCs to bacterial challenge with a wild-type strain and its isogenic mutants deficient in CPS or LPS O-antigen production. As observed by flow cytometry and confocal laser microscopy, the rate of phagocytosis was inversely proportional to the amount of CPS on the bacterial cell surface, with LPS playing little or no role. The K. pneumoniae wild-type strain induced DC maturation with upregulation of CD83, CD86, and TLR4 and downregulation of CD14 and DC-SIGN. With CPS mutants, we observed a greater decrease in DC-SIGN, suggesting a superior maturation of DCs. In addition, incubation of DCs with CPS mutants, and to a lesser extent with LPS mutants, resulted in significantly higher Th1 cytokine production. Combined, our findings suggest that K. pneumoniae CPS, by hampering bacterial binding and internalization, induces a defective immunological host response, including maturation of DCs and pro-Th1 cytokine production, whereas the LPS O antigen seems to be involved essentially in DC activation.
Collapse
|