1
|
Ni X, Liu Y, Sun M, Jiang Y, Wang Y, Ke D, Guo G, Liu K. Oral Live-Carrier Vaccine of Recombinant Lactococcus lactis Inducing Prophylactic Protective Immunity Against Helicobacter pylori Infection. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10360-x. [PMID: 39251521 DOI: 10.1007/s12602-024-10360-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Helicobacter pylori infects the gastric mucosa and induces chronic gastritis, peptic ulcers, and gastric cancer. Research has demonstrated that vaccination can induce a protective immune response and prevent H. pylori infection. Oral administration of the Lactococcus lactis live-carrier vaccine is safe and easily complied with by the public. In this study, two recombinant L. lactis strains were constructed that expressed antigens of H. pylori urease subunit alpha (UreA) and UreA fused with Escherichia coli heat-labile toxin B subunit (LTB-UreA), named LL-UreA and LL-LTB-UreA, respectively. The expression of antigen proteins was confirmed by Western blotting analysis. Survival assessment indicated that the engineered L. lactis could colonize in the digestive tract of BALB/c mice up to 10 days after the last oral administration with our immunization protocol. The ability to induce immune response and immune protective efficacy of the L. lactis were confirmed. These results indicated that oral administration with LL-UreA or LL-LTB-UreA could induce UreA-specific mucosal secretory IgA (sIgA) and cellular immune response, significantly increasing the cytokines levels of interferon-gamma (IFN-γ), interleukin (IL)-17A, and IL-10, together with the proportion of CD4+IFN-γ+ T cells and CD4+IL17A+ T cells. More importantly, oral administration of LL-UreA and LL-LTB-UreA brought about effective protection in mice to prevent H. pylori infection, especially LL-UreA, resulting in 70% of mice showing no H. pylori colonization and the remaining 30% showing only low levels of colonization. These findings underscore the potential of using orally administered engineered L. lactis vaccines to prevent H. pylori infection.
Collapse
Affiliation(s)
- Xiumei Ni
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Yu Liu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Min Sun
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Yajun Jiang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Yi Wang
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Dingxin Ke
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China
| | - Gang Guo
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China.
| | - Kaiyun Liu
- Biopharmaceutical Research Institute, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, China.
| |
Collapse
|
2
|
Oladejo M, Tijani AO, Puri A, Chablani L. Adjuvants in cutaneous vaccination: A comprehensive analysis. J Control Release 2024; 369:475-492. [PMID: 38569943 DOI: 10.1016/j.jconrel.2024.03.045] [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: 11/29/2023] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/05/2024]
Abstract
Skin is the body's largest organ and serves as a protective barrier from physical, thermal, and mechanical environmental challenges. Alongside, the skin hosts key immune system players, such as the professional antigen-presenting cells (APCs) like the Langerhans cells in the epidermis and circulating macrophages in the blood. Further, the literature supports that the APCs can be activated by antigen or vaccine delivery via multiple routes of administration through the skin. Once activated, the stimulated APCs drain to the associated lymph nodes and gain access to the lymphatic system. This further allows the APCs to engage with the adaptive immune system and activate cellular and humoral immune responses. Thus, vaccine delivery via skin offers advantages such as reliable antigen delivery, superior immunogenicity, and convenient delivery. Several preclinical and clinical studies have demonstrated the significance of vaccine delivery using various routes of administration via skin. However, such vaccines often employ adjuvant/(s), along with the antigen of interest. Adjuvants augment the immune response to a vaccine antigen and improve the therapeutic efficacy. Due to these reasons, adjuvants have been successfully used with infectious disease vaccines, cancer immunotherapy, and immune-mediated diseases. To capture these developments, this review will summarize preclinical and clinical study results of vaccine delivery via skin in the presence of adjuvants. A focused discussion regarding the FDA-approved adjuvants will address the experiences of using such adjuvant-containing vaccines. In addition, the challenges and regulatory concerns with these adjuvants will be discussed. Finally, the review will share the prospects of adjuvant-containing vaccines delivered via skin.
Collapse
Affiliation(s)
- Mariam Oladejo
- Department of Immunotherapeutics and Biotechnology, Jerry H Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN, USA.
| | - Lipika Chablani
- Wegmans School of Pharmacy, St. John Fisher University, 3690 East Ave, Rochester, NY 14618, USA.
| |
Collapse
|
3
|
Laksono BM, Sooksawasdi Na Ayudhya S, Aguilar-Bretones M, Embregts CWE, van Nierop GP, van Riel D. Human B cells and dendritic cells are susceptible and permissive to enterovirus D68 infection. mSphere 2024; 9:e0052623. [PMID: 38259063 PMCID: PMC10900886 DOI: 10.1128/msphere.00526-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Enterovirus D68 (EV-D68) is predominantly associated with mild respiratory infections, but can also cause severe respiratory disease and extra-respiratory complications, including acute flaccid myelitis. Systemic dissemination of EV-D68 is crucial for the development of extra-respiratory diseases, but it is currently unclear how EV-D68 spreads systemically (viremia). We hypothesize that immune cells contribute to the systemic dissemination of EV-D68, as this is a mechanism commonly used by other enteroviruses. Therefore, we investigated the susceptibility and permissiveness of human primary immune cells for different EV-D68 isolates. In human peripheral blood mononuclear cells inoculated with EV-D68, only B cells were susceptible but virus replication was limited. However, in B cell-rich cultures, such as Epstein-Barr virus-transformed B-lymphoblastoid cell line (BLCL) and primary lentivirus-transduced B cells, which better represent lymphoid B cells, were productively infected. Subsequently, we showed that dendritic cells (DCs), particularly immature DCs, are susceptible and permissive for EV-D68 infection and that they can spread EV-D68 to autologous BLCL. Altogether, our findings suggest that immune cells, especially B cells and DCs, could play an important role in the pathogenesis of EV-D68 infection. Infection of these cells may contribute to systemic dissemination of EV-D68, which is an essential step toward the development of extra-respiratory complications.IMPORTANCEEnterovirus D68 (EV-D68) is an emerging respiratory virus that has caused outbreaks worldwide since 2014. EV-D68 infects primarily respiratory epithelial cells resulting in mild respiratory diseases. However, EV-D68 infection is also associated with extra-respiratory complications, including polio-like paralysis. It is unclear how EV-D68 spreads systemically and infects other organs. We hypothesized that immune cells could play a role in the extra-respiratory spread of EV-D68. We showed that EV-D68 can infect and replicate in specific immune cells, that is, B cells and dendritic cells (DCs), and that virus could be transferred from DCs to B cells. Our data reveal a potential role of immune cells in the pathogenesis of EV-D68 infection. Intervention strategies that prevent EV-D68 infection of immune cells will therefore potentially prevent systemic spread of virus and thereby severe extra-respiratory complications.
Collapse
Affiliation(s)
| | | | | | | | | | - Debby van Riel
- Department of Viroscience, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
4
|
Zoe LH, David SR, Rajabalaya R. Chitosan nanoparticle toxicity: A comprehensive literature review of in vivo and in vitro assessments for medical applications. Toxicol Rep 2023; 11:83-106. [PMID: 38187113 PMCID: PMC10767636 DOI: 10.1016/j.toxrep.2023.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 01/09/2024] Open
Abstract
Topic definition This literature review aims to update the current knowledge on toxicity of chitosan nanoparticles, compare the recent findings and identify the gaps with knowledge that is present for the chitosan nanoparticles. Methods The publications between 2010 and 2020 were searched in Science Direct, Pubmed.gov, Google Scholar, Research Gate, and ClinicalTrials.gov, according to the inclusion and exclusion criteria. 30 primary research studies were obtained from the literature review to compare the in vitro in vivo toxicity profiles among the chitosan nanoparticles. Major highlights Chitosan nanoparticles and other types of nanoparticles show cytotoxic effects on cancer cells while having minimal toxicity on normal cells. This apparent effect poses some considerations for use in incorporating cancer therapeutics into chitosan nanoparticles as an administration form. The concentration, duration of exposure, and pH of the solution can influence nanoparticle cytotoxicity, particularly in zebrafish. Different cell lines exhibit varying degrees of toxicity when exposed to nanoparticles, and of note are liver cells that show toxicity under exposure as indicated by increased alanine transaminase (ALT) levels. Aside from ALT, platelet aggregation can be considered a toxicity induced by chitosan nanoparticles. In addition, zebrafish cells experience the most toxicity, including organ damage, neurobehavioral impairment, and developmental abnormalities, when exposed to nanoparticles. However, nanoparticles may exhibit different toxicity profiles in different organisms, with brain toxicity and liver toxicity being present in zebrafish but not rats. Different organs exhibit varying degrees of toxicity, with the eye and mouth apparently having the lowest toxicity, while the brain, intestine, muscles and lung showing mixed results. Cardiotoxicity induced by chitosan nanoparticles was not observed in zebrafish embryos, and nanoparticles may reduce cardiotoxicity when delivering drug. Toxicity found in an organ may not necessarily mean that it is toxic towards all the cells found in that organ, as muscle toxicity was present when tested in zebrafish but not in C2C12 myoblast cells. Some of the studies conducted may have limitations that need to be reconsidered to account for differing results, with some examples being two experiments done on HeLa cells where one study concluded chitosan nanoparticles were toxic to the cells while the other seems to have no toxicity present. With regards to LD50, one study has stated the concentration of 64.21 mg/ml was found. Finally, smaller nanoparticles generally exhibit higher toxicity in cells compared to larger nanoparticles. Scope for future work This literature review did not uncover any published clinical trials with available results. Subsequent research endeavors should prioritize conducting clinical trials involving human volunteers to directly assess toxicity, rather than relying on cell or animal models.
Collapse
Affiliation(s)
- Liaw Hui Zoe
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, BE1410 Bandar Seri Begawan, Brunei Darussalam
| | - Sheba R. David
- School of Pharmacy, University of Wyoming, Laramie, WY 82071, USA
| | - Rajan Rajabalaya
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, BE1410 Bandar Seri Begawan, Brunei Darussalam
| |
Collapse
|
5
|
Isticato R. Bacterial Spore-Based Delivery System: 20 Years of a Versatile Approach for Innovative Vaccines. Biomolecules 2023; 13:947. [PMID: 37371527 DOI: 10.3390/biom13060947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/25/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023] Open
Abstract
Mucosal vaccines offer several advantages over injectable conventional vaccines, such as the induction of adaptive immunity, with secretory IgA production at the entry site of most pathogens, and needle-less vaccinations. Despite their potential, only a few mucosal vaccines are currently used. Developing new effective mucosal vaccines strongly relies on identifying innovative antigens, efficient adjuvants, and delivery systems. Several approaches based on phages, bacteria, or nanoparticles have been proposed to deliver antigens to mucosal surfaces. Bacterial spores have also been considered antigen vehicles, and various antigens have been successfully exposed on their surface. Due to their peculiar structure, spores conjugate the advantages of live microorganisms with synthetic nanoparticles. When mucosally administered, spores expressing antigens have been shown to induce antigen-specific, protective immune responses. This review accounts for recent progress in the formulation of spore-based mucosal vaccines, describing a spore's structure, specifically the spore surface, and the diverse approaches developed to improve its efficiency as a vehicle for heterologous antigen presentation.
Collapse
Affiliation(s)
- Rachele Isticato
- Department of Biology, University of Naples Federico II, Complesso Universitario Monte S. Angelo, Via Cinthia 4, 80126 Naples, Italy
- Interuniversity Center for Studies on Bioinspired Agro-Environmental Technology (BAT Center), 80055 Naples, Italy
| |
Collapse
|
6
|
Pandey P, Kim SH, Subedi L, Mujahid K, Kim Y, Cho YC, Shim JH, Kim KT, Cho SS, Choi JU, Park JW. Oral lymphatic delivery of alpha-galactosylceramide and ovalbumin evokes anti-cancer immunization. J Control Release 2023; 356:507-524. [PMID: 36907564 DOI: 10.1016/j.jconrel.2023.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/16/2023] [Accepted: 03/06/2023] [Indexed: 03/14/2023]
Abstract
We developed an orally delivered nanoemulsion that induces cancer immunization. It consists of tumor antigen-loaded nano-vesicles carrying the potent invariant natural killer T-cell (iNKT) activator α-galactosylceramide (α-GalCer), to trigger cancer immunity by effectively activating both innate and adaptive immunity. It was validated that adding bile salts to the system boosted intestinal lymphatic transport as well as the oral bioavailability of ovalbumin (OVA) via the chylomicron pathway. To increase intestinal permeability further and amplify the antitumor responses, an ionic complex of cationic lipid 1,2-dioleyl-3-trimethylammonium propane (DTP) with sodium deoxycholate (DA) (DDP) and α-GalCer were anchored onto the outer oil layer to form OVA-NE#3. As expected, OVA-NE#3 exhibited tremendously improved intestinal cell permeability as well as enhanced delivery to mesenteric lymph nodes (MLNs). Subsequent activation of dendritic cells and iNKTs, in MLNs were also observed. Tumor growth in OVA-expressing mice with melanoma was more strongly suppressed (by 71%) after oral administration of OVA-NE#3 than in untreated controls, confirming the strong immune response induced by the system. The serum levels of OVA-specific IgG1 and IgG2a were 3.52- and 6.14-fold higher than in controls. Treating OVA-NE#3 increased the numbers of tumor-infiltrating lymphocytes, including cytotoxic T-cell and M1-like macrophage. Antigen- and α-GalCer-associated enrichment of dendritic cells and iNKTs in tumor tissues also increased after OVA-NE#3 treatment. These observations indicate that our system induces both cellular and humoral immunity by targeting the oral lymphatic system. It may offer a promising oral anti-cancer vaccination strategy that involves the induction of systemic anti-cancer immunization.
Collapse
Affiliation(s)
- Prashant Pandey
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Seung Hyun Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Laxman Subedi
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Khizra Mujahid
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yebon Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Young-Chang Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jung-Hyun Shim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Ki-Taek Kim
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Seung-Sik Cho
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jeong Uk Choi
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Jin Woo Park
- Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea; College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea.
| |
Collapse
|
7
|
Tripathi S, Sharma N, Naorem LD, Raghava GPS. ViralVacDB: A manually curated repository of viral vaccines. Drug Discov Today 2023; 28:103523. [PMID: 36764575 DOI: 10.1016/j.drudis.2023.103523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 12/13/2022] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Over the years, numerous vaccines have been developed against viral infections; however, a complete database that provides comprehensive information on viral vaccines has been lacking. In this review, along with our freely accessible database ViralVacDB, we provide details of the viral vaccines, their type, routes of administration and approving agencies. This repository systematically covers additional information such as disease name, adjuvant, manufacturer, clinical status, age and dosage against 422 viral vaccines, including 145 approved vaccines and 277 in clinical trials. We anticipate that this database will be highly beneficial to researchers and others working in pharmaceuticals and immuno-informatics.
Collapse
Affiliation(s)
- Sadhana Tripathi
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Neelam Sharma
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Leimarembi Devi Naorem
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| | - Gajendra P S Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology, Okhla Phase 3, New Delhi 110020, India.
| |
Collapse
|
8
|
Jordan-Paiz A, Martrus G, Steinert FL, Kaufmann M, Sagebiel AF, Schreurs RRCE, Rechtien A, Baumdick ME, Jung JM, Möller KJ, Wegner L, Grüttner C, Richert L, Thünauer R, Schroeder-Schwarz J, van Goudoever JB, Geijtenbeek TBH, Altfeld M, Pals ST, Perez D, Klarenbeek PL, Tomuschat C, Sauter G, Königs I, Schumacher U, Friese MA, Melling N, Reinshagen K, Bunders MJ. CXCR5 +PD-1 ++ CD4 + T cells colonize infant intestines early in life and promote B cell maturation. Cell Mol Immunol 2023; 20:201-213. [PMID: 36600048 PMCID: PMC9886971 DOI: 10.1038/s41423-022-00944-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 10/26/2022] [Indexed: 01/06/2023] Open
Abstract
Gastrointestinal infections are a major cause for serious clinical complications in infants. The induction of antibody responses by B cells is critical for protective immunity against infections and requires CXCR5+PD-1++ CD4+ T cells (TFH cells). We investigated the ontogeny of CXCR5+PD-1++ CD4+ T cells in human intestines. While CXCR5+PD-1++ CD4+ T cells were absent in fetal intestines, CXCR5+PD-1++ CD4+ T cells increased after birth and were abundant in infant intestines, resulting in significant higher numbers compared to adults. These findings were supported by scRNAseq analyses, showing increased frequencies of CD4+ T cells with a TFH gene signature in infant intestines compared to blood. Co-cultures of autologous infant intestinal CXCR5+PD-1+/-CD4+ T cells with B cells further demonstrated that infant intestinal TFH cells were able to effectively promote class switching and antibody production by B cells. Taken together, we demonstrate that functional TFH cells are numerous in infant intestines, making them a promising target for oral pediatric vaccine strategies.
Collapse
Affiliation(s)
- Ana Jordan-Paiz
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Glòria Martrus
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Fenja L Steinert
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Max Kaufmann
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Adrian F Sagebiel
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Renée R C E Schreurs
- Department of Experimental Immunology; Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Anne Rechtien
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
- Partner Site Hamburg-Lübeck-Borstel-Riems, German Center for Infection Research (DZIF), Hamburg, 20246, Germany
| | - Martin E Baumdick
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Johannes M Jung
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Kimberly J Möller
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Lucy Wegner
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
- University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Cordula Grüttner
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Laura Richert
- University of Bordeaux, Institut National de la Santé et de la Recherche Médicale, Bordeaux Population Health Research Center UMR1219 and INRIA SISTM Team, Bordeaux, 33000, France
| | - Roland Thünauer
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Jennifer Schroeder-Schwarz
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Johannes B van Goudoever
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology; Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Marcus Altfeld
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany
| | - Steven T Pals
- Department of Pathology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Daniel Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Paul L Klarenbeek
- Department of Rheumatology and Clinical Immunology and Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1007 MB, The Netherlands
- Amsterdam Rheumatology & Immunology Center, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, 1105 AZ, The Netherlands
| | - Christian Tomuschat
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Ingo Königs
- Department of Pediatric Surgery, Altona Children's Hospital, Hamburg, 22763, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, 20251, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Madeleine J Bunders
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, 20251, Germany.
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany.
| |
Collapse
|
9
|
Schwestka J, Zeh L, Tschofen M, Schubert F, Arcalis E, Esteve-Gasent M, Pedrazzini E, Vitale A, Stoger E. Generation of multi-layered protein bodies in N. benthamiana for the encapsulation of vaccine antigens. FRONTIERS IN PLANT SCIENCE 2023; 14:1109270. [PMID: 36733717 PMCID: PMC9887037 DOI: 10.3389/fpls.2023.1109270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/02/2023] [Indexed: 06/18/2023]
Abstract
The ability of plants to assemble particulate structures such as virus-like particles and protein storage organelles allows the direct bioencapsulation of recombinant proteins during the manufacturing process, which holds promise for the development of new drug delivery vehicles. Storage organelles found in plants such as protein bodies (PBs) have been successfully used as tools for accumulation and encapsulation of recombinant proteins. The fusion of sequences derived from 27-kDa-γ-zein, a major storage protein of maize, with a protein of interest leads to the incorporation of the chimeric protein into the stable and protected environment inside newly induced PBs. While this procedure has proven successful for several, but not all recombinant proteins, the aim of this study was to refine the technology by using a combination of PB-forming proteins, thereby generating multi-layered protein assemblies in N. benthamiana. We used fluorescent proteins to demonstrate that up to three proteinaceous components can be incorporated into different layers. In addition to 27-kDa-γ-zein, which is essential for PB initiation, 16-kDa-γ-zein was identified as a key element to promote the incorporation of a third zein-component into the core of the PBs. We show that a vaccine antigen could be incorporated into the matrix of multi-layered PBs, and the protein microparticles were characterized by confocal and electron microscopy as well as flow cytometry. In future, this approach will enable the generation of designer PBs that serve as drug carriers and integrate multiple components that can be functionalized in different ways.
Collapse
Affiliation(s)
- Jennifer Schwestka
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Lukas Zeh
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Marc Tschofen
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Fabian Schubert
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Elsa Arcalis
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Maria Esteve-Gasent
- Department of Veterinary Pathobiology, College of Veterinary Medicine, College Station, TX, United States
| | - Emanuela Pedrazzini
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche (CNR), Milano, Italy
| | - Alessandro Vitale
- Istituto di Biologia e Biotecnologia Agraria, Consiglio Nazionale delle Ricerche (CNR), Milano, Italy
| | - Eva Stoger
- Institute of Plant Biotechnology and Cell Biology, Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
10
|
Design, Development, and Optimisation of Smart Linker Chemistry for Targeted Colonic Delivery-In Vitro Evaluation. Pharmaceutics 2023; 15:pharmaceutics15010303. [PMID: 36678931 PMCID: PMC9860859 DOI: 10.3390/pharmaceutics15010303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
Drug targeting is necessary to deliver drugs to a specific site of action at a rate dictated by therapeutic requirements. The pharmacological action of a drug can thereby be optimised while minimising adverse effects. Numerous colonic drug delivery systems have been developed to avoid such undesirable side effects; however, these systems lack site specificity, leaving room for further improvement. The objective of the present study was to explore the potential of amino-alkoxycarbonyloxymethyl (amino-AOCOM) ether prodrugs as a general approach for future colonic delivery. To circumvent inter- and intra-subject variabilities in enzyme activities, these prodrugs do not rely on enzymes but rather are activated via a pH-triggered intramolecular cyclisation−elimination reaction. As proof of concept, model compounds were synthesised and evaluated under various pH conditions, simulating various regions of the gastrointestinal tract (GIT). Probe 15 demonstrated excellent stability under simulated stomach- and duodenum-like conditions and protected 60% of the payload in a small intestine-like environment. Moreover, 15 displayed sustained release at colonic pH, delivering >90% of the payload over 38 h. Mesalamine (Msl) prodrugs 21 and 22 were also synthesised and showed better stability than probe 15 in the simulated upper GIT but relatively slower release at colonic pH (61−68% of Msl over 48 h). For both prodrugs, the extent of release was comparable to that of the commercial product Asacol. This study provides initial proof of concept regarding the use of a cyclisation-activated prodrug for colon delivery and suggests that release characteristics still vary on a case-by-case basis.
Collapse
|
11
|
Gupta P, Andankar I, Gunasekaran B, Easwaran N, Kodiveri Muthukaliannan G. Genetically modified potato and rice based edible vaccines – An overview. BIOCATALYSIS AND AGRICULTURAL BIOTECHNOLOGY 2022. [DOI: 10.1016/j.bcab.2022.102405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
12
|
Guo L, Zhang F, Wang S, Li R, Zhang L, Zhang Z, Yin R, Liu H, Liu K. Oral Immunization With a M Cell-Targeting Recombinant L. Lactis Vaccine LL-plSAM-FVpE Stimulate Protective Immunity Against H. Pylori in Mice. Front Immunol 2022; 13:918160. [PMID: 35911756 PMCID: PMC9336465 DOI: 10.3389/fimmu.2022.918160] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/15/2022] [Indexed: 11/24/2022] Open
Abstract
There are many virulence factors of H. pylori that contribute in diverse ways to gastric disease. Therefore, designing multivalent epitope vaccines against many key virulence factors virulence factors of H. pylori is a promising strategy to control H. pylori infection. In previous studies, we constructed a multivalent epitope vaccine FVpE against four key virulence factors of H. pylori (Urease, CagA, VacA, and NAP), and oral immunization with the FVpE vaccine plus a polysaccharide adjuvant (PA) containing lycium barbarum polysaccharide and chitosan could provide protection against H. pylori infection in the Mongolian gerbil model. Oral vaccines have many advantages over injected vaccines, such as improved safety and compliance, and easier manufacturing and administration. However, the harsh gastrointestinal (GI) environment, such as gastric acid and proteolytic enzymes, limits the development of oral vaccines to some extent. Oral vaccines need a gastrointestinal delivery system with high safety, low price and promoting vaccine antigen to stimulate immune response in the gastrointestinal mucosa. Lactic acid bacteria are gastrointestinal probiotics that have unique advantages as a delivery system for oral vaccines. In this study, a M cell-targeting surface display system for L. lactis named plSAM was designed to help vaccine antigens to stimulate effective immune responses in the gastrointestinal tract, and a M cell-targeting recombinant L. lactis vaccine LL-plSAM-FVpE was constructed by using the surface display system plSAM. recombinant L. lactis vaccine LL-plSAM-FVpE could secretively express the SAM-FVpE protein and display it on the bacterial surface. Moreover, experimental results confirmed that LL-plSAM-FVpE had an enhanced M cell-targeting property. In addition, LL-plSAM-FVpE had excellent M cell-targeting property to promote the phagocytosis and transport of the antigen SAM-FVpE by gastrointestinal M cells. More importantly, oral immunization of LL-plSAM-FVpE or SAM-FVpE plus PA can stimulate IgG and sIgA antibodies and CD4+ T cell immune responses against four virulence factors of H. pylori (Urease, CagA, VacA, and NAP), thus providing protective immunity against H. pylori infection in mice. The M cell-targeting recombinant L. lactis vaccine against various key H. pylori virulence factors could be a promising vaccine candidate for controlling H. pylori infection.
Collapse
Affiliation(s)
- Le Guo
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
- Key Laboratory of Radiation Oncology of Taizhou, Taizhou Hospital of Zhejiang Province affifiliated to Wenzhou Medical University, Taizhou, China
| | - Furui Zhang
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Shue Wang
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Runle Li
- Research Center for High Altitude Medicine, Qinghai University, Xining, China
| | - Lele Zhang
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Zhen Zhang
- Cancer Hospital, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Runting Yin
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Hongpeng Liu
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- *Correspondence: Kunmei Liu, ; Hongpeng Liu,
| | - Kunmei Liu
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan, China
- *Correspondence: Kunmei Liu, ; Hongpeng Liu,
| |
Collapse
|
13
|
Angulo C, Sanchez V, Delgado K, Monreal-Escalante E, Hernández-Adame L, Angulo M, Tello-Olea M, Reyes-Becerril M. Oral organic nanovaccines against bacterial and viral diseases. Microb Pathog 2022; 169:105648. [PMID: 35728750 DOI: 10.1016/j.micpath.2022.105648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/12/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023]
Abstract
Vaccines have saved millions of humans and animals from deadly diseases. Many vaccines are still under development to fight against lethal diseases. Indeed, subunit vaccines are a versatile approach with several advantageous attributes, but they lack strong immunogenicity. Nanotechnology is an avenue to vaccine development because nanoparticles may serve as nanocarriers and adjuvants, which are critical aspects for oral vaccines. This review provides an update of oral organic nanovaccines, describing suitable nanomaterials for oral vaccine design and recent (last five-year view) oral nanovaccine developments to fight against those principal pathogens causing human and animal diseases.
Collapse
Affiliation(s)
- Carlos Angulo
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico.
| | - Veronica Sanchez
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Karen Delgado
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Elizabeth Monreal-Escalante
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico; Cátedras-CONACYT. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Luis Hernández-Adame
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico; Cátedras-CONACYT. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Miriam Angulo
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Marlene Tello-Olea
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| | - Martha Reyes-Becerril
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S., 23096, Mexico
| |
Collapse
|
14
|
Zhang L, Yao L, Guo Y, Li X, Ma L, Sun R, Han X, Liu J, Huang J. Oral SARS-CoV-2 Spike Protein Recombinant Yeast Candidate Prompts Specific Antibody and Gut Microbiota Reconstruction in Mice. Front Microbiol 2022; 13:792532. [PMID: 35464985 PMCID: PMC9022078 DOI: 10.3389/fmicb.2022.792532] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
A recent study showed that patients with coronavirus disease 2019 (COVID-19) have gastrointestinal symptoms and intestinal flora dysbiosis. Yeast probiotics shape the gut microbiome and improve immune homeostasis. In this study, an oral candidate of yeast-derived spike protein receptor-binding domain (RBD) and fusion peptide displayed on the surface of the yeast cell wall was generated. The toxicity and immune efficacy of oral administration were further performed in Institute of Cancer Research (ICR) mice. No significant difference in body weights, viscera index, and other side effects were detected in the oral-treated group. The detectable RBD-specific immunoglobulin G (IgG) and immunoglobulin A (IgA) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and more complex microbiota were detected from oral administration mice compared with those of the control group. Interestingly, the recombinant yeast was identified in female fetal of the high-dose group. These results revealed that the displaying yeast could fulfill the agent-driven immunoregulation and gut microbiome reconstitution. The findings will shed light on new dimensions against SARS-CoV-2 infection with the synergistic oral agents as promising non-invasive immunization and restoring gut flora.
Collapse
Affiliation(s)
- Lilin Zhang
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Lan Yao
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Yanyu Guo
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Xiaoyang Li
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Li Ma
- Tianjin Institute of Pharmaceutical Research Co., Ltd., Tianjin, China
| | - Ruiqi Sun
- School of Life Sciences, Tianjin University, Tianjin, China
| | - Xueqing Han
- Chinese Academy of Inspection and Quarantine, Beijing, China
| | - Jing Liu
- Tianjin Institute of Pharmaceutical Research Co., Ltd., Tianjin, China
- Jing Liu,
| | - Jinhai Huang
- School of Life Sciences, Tianjin University, Tianjin, China
- *Correspondence: Jinhai Huang,
| |
Collapse
|
15
|
Advancedoral vaccine delivery strategies for improving the immunity. Adv Drug Deliv Rev 2021; 177:113928. [PMID: 34411689 DOI: 10.1016/j.addr.2021.113928] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/15/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022]
Abstract
Infectious diseases continue to inflict a high global disease burden. The consensus is that vaccination is the most effective option against infectious diseases. Oral vaccines have unique advantages in the prevention of global pandemics due to their ease of use, high compliance, low cost, and the ability to induce both systemic and mucosal immune responses. However, challenges of adapting vaccines for oral administration remain significant. Foremost among these are enzymatic and pH-dependent degradation of antigens in the stomach and intestines, the low permeability of mucus barrier, the nonspecific uptake of antigens at the intestinal mucosal site, and the immune suppression result from the elusive immune tolerance mechanisms. Innovative delivery techniques promise great potential for improving the flexibility and efficiency of oral vaccines. A better understanding of the delivery approaches and the immunological mechanisms of oral vaccine delivery systems may provide new scientific insight and tools for developing the next-generation oral vaccine. Here, an overview of the advanced technologies in the field of oral vaccination is proposed, including mucus-penetrating nanoparticle (NP), mucoadhesive delivery vehicles, targeting antigen-presenting cell (APC) nanocarriers and enhanced paracellular delivery strategies and so on. Meanwhile, the mechanisms of delivery vectors interact with mucosal barriers are discussed.
Collapse
|
16
|
Microparticles and Nanoparticles from Plants-The Benefits of Bioencapsulation. Vaccines (Basel) 2021; 9:vaccines9040369. [PMID: 33920425 PMCID: PMC8069552 DOI: 10.3390/vaccines9040369] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/04/2021] [Accepted: 04/09/2021] [Indexed: 11/25/2022] Open
Abstract
The efficacy of drugs and vaccines depends on their stability and ability to interact with their targets in vivo. Many drugs benefit from encapsulation, which protects them from harsh conditions and allows targeted delivery and controlled release. Although many encapsulation methods are inexpensive, such as the formulation of tablets for oral delivery, others require complex procedures that add significantly to production costs and require low-temperature transport and storage, making them inaccessible in developing countries. In this review we consider the benefits of encapsulation technologies based on plants. Plant-derived biopolymers such as starch and the maize storage protein zein are already used as protective coatings, but plant cells used as production host provide natural in vivo bioencapsulation that survives passage through the stomach and releases drugs in the intestine, due to the presence of microbes that can digest the cell wall. Proteins can also be encapsulated in subcellular compartments such as protein bodies, which ensure stability and activity while often conferring additional immunomodulatory effects. Finally, we consider the incorporation of drugs and vaccines into plant-derived nanoparticles assembled from the components of viruses. These are extremely versatile, allowing the display of epitopes and targeting peptides as well as carrying cargoes of drugs and imaging molecules.
Collapse
|
17
|
Hewawaduge C, Senevirathne A, Yang MS, Jeong TW, Kim B, Lee JH. Comparative study of sodium bicarbonate- and magnesium hydroxide-based gastric antacids for the effectiveness of Salmonella delivered Brucella antigens against wild type challenge in BALB/c mice. Pathog Dis 2021; 79:6126344. [PMID: 33527985 DOI: 10.1093/femspd/ftab002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/11/2021] [Indexed: 11/14/2022] Open
Abstract
We compared the effects of two antacid formulations based on sodium bicarbonate and magnesium hydroxide on a Salmonella-delivered oral Brucella live attenuated vaccine. We conducted a series of in vitro and in vivo experiments to investigate the pH buffering capacity, buffering longevity and the effects of these formulations on the survival of Salmonella under neutralized pH conditions and its impact on immune responses. Magnesium hydroxide had a greater, stable and prolonged buffering capacity than sodium bicarbonate and was safer when administered orally. Oral administration of sodium bicarbonate resulted in discomfort as reflected by mouse behavior and mild muscle tremors, whereas mice treated with magnesium hydroxide and PBS were completely normal. Gastric survival studies using BALB/c mice revealed that a higher number of Salmonella reached the intestine when the magnesium hydroxide-based antacid buffer was administrated. Co-administration with attenuated Salmonella secreting Brucella antigens, SodC and Omp19 along with individual antacid formulations, significantly enhanced the antigen-specific protective immune responses against virulent Brucella challenge. Together, our results indicated that the pre vaccinated oral administration of bicarbonate-citric acid or magnesium hydroxide-based neutralizing buffers significantly counteract stomach acidity by maintaining the viability of an oral enteric vaccine formulation.
Collapse
Affiliation(s)
- Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Amal Senevirathne
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Myeon-Sik Yang
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Tae-Won Jeong
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - Bumseok Kim
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus, 54596, Iksan, Republic of Korea
| |
Collapse
|
18
|
Li JB, Li JJ, Li M, Gao C, Zhang L, Li M, Zhu Q. Oral immunization induces a novel CXCR6 + β7 + intraepithelial lymphocyte subset predominating in the small intestine. Scand J Immunol 2020; 93:e12996. [PMID: 33205443 DOI: 10.1111/sji.12996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/27/2022]
Abstract
Intestinal T cells form a central part of the front-line defence against foreign organisms and need to be situated in the mucosa where infection occurs. It is well accepted that immunization by a mucosal route favours localization of antigen-specific effector T cells in the mucosal epithelium, while systemic immunization does not. The aim of the study is to determine how homing receptors are specifically involved in retaining effector T cells in the small intestine after oral immunization. We here demonstrate that the chemokine receptor CXCR6, integrins β7 and CD29 contribute differentially to the epithelial retention phenotype of CD8+ T cells in the small intestine of mice. CD8+ intraepithelial lymphocytes (IELs) of unvaccinated mice are predominantly β7 single positives, and subcutaneous immunization-induced antigen-specific CD8+ effector IELs are mainly composed of CXCR6+ , CD29+ and CXCR6+ CD29+ cells. Strikingly, the majority of oral immunization-induced antigen-specific CD8+ effector IELs exhibit a distinct, tissue-specific CXCR6+ β7+ double-positive phenotype, cytotoxic potential and enhanced intraepithelial localization. Transfer of antigen-specific CD8+ T cells preactivated with certain immuno-stimuli (such as monophosphoryl lipid A) results in increased accumulation of donor IELs with the CXCR6+ β7+ phenotype. As β7 exclusively paired with αE on IELs, our results strongly suggest that CXCR6 may cooperate with the heterodimer αEβ7 to preferentially retain intestinally induced effector IELs in the epithelium. The identification of this novel IEL phenotype has significant implications for the development of vaccines and therapeutic strategies to enhance gut immunity.
Collapse
Affiliation(s)
- Jing B Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing, China
| | - Jing J Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing, China
| | - Mingyan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing, China
| | - Changxing Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing, China
| | - Lingzhi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing, China
| | - Meihan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing, China
| | - Qing Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing, China
| |
Collapse
|
19
|
Saraf S, Jain S, Sahoo RN, Mallick S. Present Scenario of M-Cell Targeting Ligands for Oral Mucosal Immunization. Curr Drug Targets 2020; 21:1276-1284. [DOI: 10.2174/1389450121666200609113252] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/18/2020] [Accepted: 04/23/2020] [Indexed: 02/07/2023]
Abstract
The immune system plays an important role in the prevention of infection and forms the
first line of defense against pathogen attack. Delivering of antigen through mucosal route may elicit
mucosal immune system as the mucosal surface is the most common site of pathogen entry. Mucosal
immune system will be capable to counter pathogen at mucosal surface. Oral mucosal immunization
opens the ways to deliver antigens at gut-associated lymphoid tissue. This can elicit both local and
systemic immune response. Mucosal vaccines are economical, highly accessible, non parenteral delivery
and capacity to produce mass immunization at the time of pandemics. To deliver antigens on the
mucosal surface of the gastrointestinal tract, the immune system relies on specialized epithelial cell
i.e. Microfold (M)-cell. An approach to exploit the targeting specific receptors on M-cell for entry of
antigens has made a breakthrough in vaccine development. In this review, various strategies have been
discussed for the possible entry of antigens through M-cells and an approach to increase the uptake
and efficacy of vaccines for oral mucosal immunization.
Collapse
Affiliation(s)
- Surendra Saraf
- School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751030, Orissa, India
| | - Shailesh Jain
- Dean, Faculty of Pharmacy and Pharmaceutical Sciences at Madhyanchal Professional University Bhopal (MP), India
| | - Rudra Narayan Sahoo
- School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751030, Orissa, India
| | - Subrata Mallick
- School of Pharmaceutical Sciences, Siksha ‘O’ Anusandhan (Deemed to be University), Bhubaneswar-751030, Orissa, India
| |
Collapse
|
20
|
Basak S, Chu KB, Kang HJ, Kim MJ, Lee SH, Yoon KW, Jin H, Suh JW, Moon EK, Quan FS. Orally administered recombinant baculovirus vaccine elicits partial protection against avian influenza virus infection in mice. Microb Pathog 2020; 149:104495. [PMID: 32910984 DOI: 10.1016/j.micpath.2020.104495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/20/2020] [Accepted: 08/31/2020] [Indexed: 12/23/2022]
Abstract
Avian influenza outbreaks have placed a tremendous economic burden on the poultry industry, necessitating the need for an effective vaccine. Although multiple vaccine candidates are available, its development is hindered by several drawbacks associated with the vaccine platforms and as such, more improvements to the vaccines are needed. Therefore, in this study, the vaccine efficacy in the murine models was assessed prior to evaluation in chickens. An oral recombinant baculovirus (rBV) vaccine expressing influenza hemagglutinin (HA) (A/H5N1) was generated and its efficacy was investigated against homologous avian influenza infection in mice. Our results confirmed that oral administration of rBVs enhanced the level of virus-specific antibodies in the sera following boost immunization. Upon challenge infection with a lethal dose of highly pathogenic avian influenza virus (HPAI, H5N1) virus, a marked increase in mucosal IgG and IgA were observed. Drastically increased antibody secretory cell responses from the bone marrow cells and splenocytes of vaccinated mice were observed, in addition to the strongly elicited germinal center responses in the lungs and the spleens. Vaccinated mice showed significantly reduced lung pro-inflammatory cytokine responses, lung viral loads, body weight loss, and mortality. Though mice were only partially protected upon challenge infection, these results highlight the potential of orally administered rBVs expressing the HA as a vaccine candidate for controlling avian influenza outbreaks.
Collapse
Affiliation(s)
- Swarnendu Basak
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Ki-Back Chu
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hae-Ji Kang
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Su-Hwa Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hui Jin
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Gyeonggi-do, Republic of Korea
| | - Joo Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Gyeonggi-do, Republic of Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul, Republic of Korea; Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
21
|
Nahm MH, Yu J, Vlach J, Bar-Peled M. A Common Food Glycan, Pectin, Shares an Antigen with Streptococcus pneumoniae Capsule. mSphere 2020; 5:e00074-20. [PMID: 32269150 PMCID: PMC7142292 DOI: 10.1128/msphere.00074-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/20/2020] [Indexed: 01/31/2023] Open
Abstract
We are exposed daily to many glycans from bacteria and food plants. Bacterial glycans are generally antigenic and elicit antibody responses. It is unclear if food glycans' sharing of antigens with bacterial glycans influences our immune responses to bacteria. We studied 14 different plant foods for cross-reactivity with monoclonal antibodies (MAbs) against 24 pneumococcal serotypes which commonly cause infections and are included in pneumococcal vaccines. Serotype 15B-specific MAb cross-reacts with fruit peels, and serotype 10A MAb cross-reacts with many natural and processed plant foods. The serotype 10A cross-reactive epitope is terminal 1,6-linked β-galactose [βGal(1-6)], present in the rhamno-galacturonan I (RG-I) domain of pectin. Despite wide consumption of pectin, the immune response to 10A is comparable to the responses to other serotypes. An antipectin antibody can opsonize serotype 10A pneumococci, and the shared βGal(1-6) may be useful as a simple vaccine against 10A. Impact of food glycans should be considered in host-pathogen interactions and future vaccine designs.IMPORTANCE The impact of food consumption on vaccine responses is unknown. Streptococcus pneumoniae (the pneumococcus) is an important human pathogen, and its polysaccharide capsule is used as a vaccine. We show that capsule type 10A in a pneumococcal vaccine shares an antigenic epitope, βGal(1-6), with pectin, which is in many plant foods and is widely consumed. Immune response to 10A is comparable to that seen with other capsule types, and pectin ingestion may have little impact on vaccine responses. However, antibody to pectin can kill serotype 10A pneumococci and this shared epitope may be considered in pneumococcal vaccine designs.
Collapse
Affiliation(s)
- Moon H Nahm
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jigui Yu
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jiri Vlach
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Maor Bar-Peled
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
22
|
Van der Weken H, Cox E, Devriendt B. Rapid production of a chimeric antibody-antigen fusion protein based on 2A-peptide cleavage and green fluorescent protein expression in CHO cells. MAbs 2019; 11:559-568. [PMID: 30694096 PMCID: PMC6512901 DOI: 10.1080/19420862.2019.1574531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/04/2019] [Accepted: 01/21/2019] [Indexed: 01/22/2023] Open
Abstract
To enable large-scale antibody production, the creation of a stable, high producer cell line is essential. This process often takes longer than 6 months using standard limited dilution techniques and is very labor intensive. The use of a tri-cistronic vector expressing green fluorescent protein (GFP) and both antibody chains, separated by a GT2A peptide sequence, allows expression of all proteins under a single promotor in equimolar ratios. By combining the advantages of 2A peptide cleavage and single cell sorting, a chimeric antibody-antigen fusion protein that contained the variable domains of mouse IgG with a porcine IgA constant domain fused to the FedF antigen could be produced in CHO-K1 cells. After transfection, a strong correlation was found between antibody production and GFP expression (r = 0.69) using image analysis of formed monolayer patches. This enables the rapid selection of GFP-positive clones using automated image analysis for the selection of high producer clones. This vector design allowed the rapid selection of high producer clones within a time-frame of 4 weeks after transfection. The highest producing clone had a specific antibody productivity of 2.32 pg/cell/day. Concentrations of 34 mg/L were obtained using shake-flask batch culture. The produced recombinant antibody showed stable expression, binding and minimal degradation. In the future, this antibody will be assessed for its effectiveness as an oral vaccine antigen.
Collapse
Affiliation(s)
- Hans Van der Weken
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ughent, Ghent, Belgium
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ughent, Ghent, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ughent, Ghent, Belgium
| |
Collapse
|
23
|
Diaz-Dinamarca DA, Soto DA, Leyton YY, Altamirano-Lagos MJ, Avendaño MJ, Kalergis AM, Vasquez AE. Oral vaccine based on a surface immunogenic protein mixed with alum promotes a decrease in Streptococcus agalactiae vaginal colonization in a mouse model. Mol Immunol 2018; 103:63-70. [PMID: 30205305 DOI: 10.1016/j.molimm.2018.08.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/07/2018] [Accepted: 08/31/2018] [Indexed: 01/15/2023]
Abstract
The Surface Immunogenic Protein (SIP) of Group B Streptococcus (GBS) had been described as a good target for vaccine development. To date, SIP has been reported as a highly conserved protein, and in a mouse model it induces protection against lethal GBS challenge. Also, similar effects have been described by intranasal immunization with a SIP-based vaccine. In this study, we show the immune response induced by an oral SIP-based vaccine formulated on alum in a mouse model. Our vaccine can reduce vaginal GBS colonization and induce specific SIP-antibodies with opsonophagocytosis activities against GBS. Moreover, we observed the activation of T-cells producing IFN-γ, TNF-α, IL-10, IL-2, and increased expression of the transcription factor T-bet, suggesting a Th1-type humoral response. The oral SIP-based vaccine is a novel alternative in the development of a vaccine against GBS.
Collapse
Affiliation(s)
- D A Diaz-Dinamarca
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile; Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - D A Soto
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile
| | - Y Y Leyton
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile
| | - M J Altamirano-Lagos
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile; Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M J Avendaño
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile; Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A M Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A E Vasquez
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile; Universidad San Sebastián, Facultad de Medicina y Ciencia, Escuela de Bioquímica, Providencia, Santiago, Chile.
| |
Collapse
|
24
|
Premanand B, Zhong Wee P, Prabakaran M. Baculovirus Surface Display of Immunogenic Proteins for Vaccine Development. Viruses 2018; 10:E298. [PMID: 29857561 PMCID: PMC6024371 DOI: 10.3390/v10060298] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/22/2018] [Accepted: 05/28/2018] [Indexed: 12/25/2022] Open
Abstract
Vaccination is an efficient way to prevent the occurrence of many infectious diseases in humans. To date, several viral vectors have been utilized for the generation of vaccines. Among them, baculovirus-categorized as a nonhuman viral vector-has been used in wider applications. Its versatile features, like large cloning capacity, nonreplicative nature in mammalian cells, and broad tissue tropism, hold it at an excellent position among vaccine vectors. In addition to ease and safety during swift production, recent key improvements to existing baculovirus vectors (such as inclusion of hybrid promoters, immunostimulatory elements, etc.) have led to significant improvements in immunogenicity and efficacy of surface-displayed antigens. Furthermore, some promising preclinical results have been reported that mirror the scope and practicality of baculovirus as a vaccine vector for human applications in the near future. Herein, this review provides an overview of the induced immune responses by baculovirus surface-displayed vaccines against influenza and other infectious diseases in animal models, and highlights the strategies applied to enhance the protective immune responses against the displayed antigens.
Collapse
Affiliation(s)
- Balraj Premanand
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore.
| | - Poh Zhong Wee
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore.
| | - Mookkan Prabakaran
- Temasek Life Sciences Laboratory, 1 Research Link, National University of Singapore, Singapore 117604, Singapore.
| |
Collapse
|
25
|
Bagherpour G, Ghasemi H, Zand B, Zarei N, Roohvand F, Ardakani EM, Azizi M, Khalaj V. Oral Administration of Recombinant Saccharomyces boulardii Expressing Ovalbumin-CPE Fusion Protein Induces Antibody Response in Mice. Front Microbiol 2018; 9:723. [PMID: 29706942 PMCID: PMC5908956 DOI: 10.3389/fmicb.2018.00723] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/27/2018] [Indexed: 12/22/2022] Open
Abstract
Saccharomyces boulardii, a subspecies of Saccharomyces cerevisiae, is a well-known eukaryotic probiotic with many benefits for human health. In the present study, a recombinant strain of S. boulardii was prepared to use as a potential oral vaccine delivery vehicle. In this sense, a ura3 auxotroph strain of S. boulardii CNCM I-745 (known as S. cerevisiae HANSEN CBS 5926, Yomogi®) was generated using CRISPR/Cas9 methodology. Then a gene construct encoding a highly immunogenic protein, ovalbumin (OVA), was prepared and transformed into the ura3- S. boulardii. To facilitate the transport of the recombinant immunogen across the intestinal barrier, a claudin-targeting sequence from Clostridium perfringens enterotoxin (CPE) was added to the C-terminus of the expression cassette. The recombinant S. boulardii strain expressing the OVA-CPE fusion protein was then administered orally to a group of mice, and serum IgG and fecal IgA levels were evaluated by ELISA. Our results demonstrated that anti-OVA IgG in serum significantly increased in test group (P < 0.001) compared to control groups (receiving wild type S. boulardii or PBS), and the fecal IgA titer was significantly higher in test group (P < 0.05) than control groups. In parallel, a recombinant S. boulardii strain expressing the similar construct lacking C-terminal CPE was also administered orally. The result showed an increased level of serum IgG in group receiving yeasts expressing the CPE negative construct compared to control groups; however, the fecal IgA levels did not increase significantly. In conclusion, our findings indicated that the yeast S. boulardii, as a delivery vehicle with possible immunomodulatory effects, and c-CPE, as a targeting tag, synergistically assist to stimulate systemic and local immunity. This proposed recombinant S. boulardii system might be useful in the expression of other antigenic peptides, making it as a promising tool for oral delivery of vaccines or therapeutic proteins.
Collapse
Affiliation(s)
- Ghasem Bagherpour
- Department of Medical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Hosnie Ghasemi
- Department of Medical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Bahare Zand
- Department of Medical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Najmeh Zarei
- Department of Medical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Farzin Roohvand
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Esmat M Ardakani
- Department of Molecular Medicine, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Azizi
- Department of Medical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Vahid Khalaj
- Department of Medical Biotechnology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
26
|
Abstract
Salmonella enterica subspecies enterica includes several serovars infecting both humans and other animals and leading to typhoid fever or gastroenteritis. The high prevalence of associated morbidity and mortality, together with an increased emergence of multidrug-resistant strains, is a current global health issue that has prompted the development of vaccination strategies that confer protection against most serovars. Currently available systemic vaccine approaches have major limitations, including a reduced effectiveness in young children and a lack of cross-protection among different strains. Having studied host-pathogen interactions, microbiologists and immunologists argue in favor of topical gastrointestinal administration for improvement in vaccine efficacy. Here, recent advances in this field are summarized, including mechanisms of bacterial uptake at the intestinal epithelium, the assessment of protective host immunity, and improved animal models that closely mimic infection in humans. The pros and cons of existing vaccines are presented, along with recent progress made with novel formulations. Finally, new candidate antigens and their relevance in the refined design of anti-Salmonella vaccines are discussed, along with antigen vectorization strategies such as nanoparticles or secretory immunoglobulins, with a focus on potentiating mucosal vaccine efficacy.
Collapse
|
27
|
Kammona O, Bourganis V, Karamanidou T, Kiparissides C. Recent developments in nanocarrier-aided mucosal vaccination. Nanomedicine (Lond) 2017; 12:1057-1074. [PMID: 28440707 DOI: 10.2217/nnm-2017-0015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To date, most of the licensed vaccines for mucosal delivery are based on live-attenuated viruses which carry the risk of regaining their pathogenicity. Therefore, the development of efficient nonviral vectors allowing the induction of potent humoral and cell-mediated immunity is regarded as an imperative scientific challenge as well as a commercial breakthrough for the pharma industries. For a successful translation to the clinic, such nanocarriers should protect the antigens from mucosal enzymes, facilitate antigen uptake by microfold cells and allow the copresentation of robust, safe for human use, mucosal adjuvants to antigen-presenting cells. Finally, the developed formulations should exhibit accuracy regarding the administered dose, a major drawback of mucosal vaccines in comparison with parenteral ones.
Collapse
Affiliation(s)
- Olga Kammona
- Chemical Process & Energy Resources Institute, Centre for Research & Technology Hellas, PO Box 60361, 57001 Thessaloniki, Greece
| | - Vassilis Bourganis
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece
| | - Theodora Karamanidou
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece
| | - Costas Kiparissides
- Department of Chemical Engineering, Aristotle University of Thessaloniki, PO Box 472, 54124 Thessaloniki, Greece.,Chemical Process & Energy Resources Institute, Centre for Research & Technology Hellas, PO Box 60361, 57001 Thessaloniki, Greece
| |
Collapse
|
28
|
Vaccine Delivery. Drug Deliv 2016. [DOI: 10.1201/9781315382579-18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
29
|
Aryamvally A, Gunasekaran V, Narenthiran KR, Pasupathi R. New Strategies Toward Edible Vaccines: An Overview. J Diet Suppl 2016; 14:101-116. [PMID: 27065206 DOI: 10.3109/19390211.2016.1168904] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
With the ever growing population, advancements in edible vaccines and related technologies have seen a rise in popularity. Antigenic peptides incorporated into an edible part of a plant can be administered raw as a vaccine. While conventional vaccines have improved the quality of life by drastically reducing the onset of diseases, edible vaccines are able to perform the same with greater accessibility and at an affordable price. Low cost of production, ease of storage, transportation and administration are some of the many reasons behind the push for the development of edible vaccines. This article aims at giving an overview of the different plant systems used to produce vaccines in various experiments, as well as the merits and demerits of using that particular expression system. Further, the article elaborates on the problems faced in the production of edible vaccines and the measures adopted to surpass them. The major obstacle in the process is attaining a sufficiently large concentration of foreign antigen in the plant system. The article discusses various plant expression systems like banana, rice, alfalfa, mushroom, potato, tomato, pea, tobacco, and maize. When these were reviewed, it was found that the inability to produce the desired antigen concentration was one of the primary reasons why edible vaccines sometimes fail to generate the desired level of immune response in the recipient. We conclude with a promising solution to the problem by incorporating nano-technological advancements to the already existing protocols for edible vaccine development.
Collapse
Affiliation(s)
- Anjali Aryamvally
- a Department of Genetic Engineering , SRM University , Kattankulathur , Tamil Nadu , India
| | - Vignesh Gunasekaran
- a Department of Genetic Engineering , SRM University , Kattankulathur , Tamil Nadu , India
| | | | | |
Collapse
|
30
|
Snook JD, Chesson CB, Peniche AG, Dann SM, Paulucci A, Pinchuk IV, Rudra JS. Peptide nanofiber–CaCO3 composite microparticles as adjuvant-free oral vaccine delivery vehicles. J Mater Chem B 2016; 4:1640-1649. [DOI: 10.1039/c5tb01623a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
To combat mucosal pathogens that cause gastrointestinal (GI) infections, local mucosal immunity is required which is best achieved through oral vaccination.
Collapse
Affiliation(s)
- Joshua D. Snook
- Department of Pharmacology & Toxicology
- University of Texas Medical Branch
- Galveston
- USA
| | - Charles B. Chesson
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Sealy Center for Vaccine Development
| | - Alex G. Peniche
- Department of Internal Medicine-Division of Infectious Diseases
- University of Texas Medical Branch
- Galveston
- USA
| | - Sara M. Dann
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Department of Internal Medicine-Division of Infectious Diseases
| | | | - Iryna V. Pinchuk
- Institute for Translation Sciences
- University of Texas Medical Branch
- Galveston
- USA
- Department of Internal Medicine-Division of Gastroenterology
| | - Jai S. Rudra
- Department of Pharmacology & Toxicology
- University of Texas Medical Branch
- Galveston
- USA
- Sealy Center for Vaccine Development
| |
Collapse
|
31
|
Biswas S, Chattopadhyay M, Sen KK, Saha MK. Development and characterization of alginate coated low molecular weight chitosan nanoparticles as new carriers for oral vaccine delivery in mice. Carbohydr Polym 2015; 121:403-10. [DOI: 10.1016/j.carbpol.2014.12.044] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 12/09/2014] [Accepted: 12/10/2014] [Indexed: 12/29/2022]
|