1
|
Tran TH, Le TH, Tran TTP. The Potential Effect of Endogenous Antimicrobial Peptides in Cancer Immunotherapy and Prevention. J Pept Sci 2025; 31:e3664. [PMID: 39716371 DOI: 10.1002/psc.3664] [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: 09/04/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 12/25/2024]
Abstract
Antimicrobial peptides (AMPs) are crucial constituents of inherent immunity and serve as vital components of human host defense, playing a pivotal role in combating invading microbial pathogens. Beyond their antimicrobial functions, AMPs also exhibit various other biological activities including apoptosis induction, wound healing promotion, and immune modulation. These peptides are found in various exposed tissues or surfaces throughout the body, such as eyes, skin, mouth, ears, respiratory tract, lungs, digestive, and urinary system. Additionally, certain AMPs such as LL-37, HNP, and lactoferrin have shown potential as candidates for anticancer activity. Given the limited selectivity between normal and cancer cells exhibited by many current immunotherapeutic agents, the inherent properties of AMPs make them promising candidates for cancer treatment. Their abundance, bioavailability, safety profile, efficiency, and harmony with the host immune system position them as attractive tools in the fight against cancer. This review is aimed at exploring the potential anticancer properties of AMPs and elucidating their relationship with immunology and cancer immunotherapy.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Faculty of Pharmacy, Phenikaa University, Hanoi, Vietnam
| | - Thanh Huong Le
- University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Thi Thu Phuong Tran
- University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| |
Collapse
|
2
|
Ding J, Zhang SS, Fernandopulle NA, Karas JA, Li J, Ziogas J, Velkov T, Mackay GA. Differential MRGPRX2-dependent activation of human mast cells by polymyxins and octapeptins. Eur J Pharmacol 2024; 984:177050. [PMID: 39389528 DOI: 10.1016/j.ejphar.2024.177050] [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: 06/30/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The emergence of multi-drug resistant Gram-negative bacteria has led to renewed interest in the antimicrobial activity of polymyxins and novel polymyxin analogues (e.g. nonapeptides and octapeptin). In some individuals, clinically used polymyxins can cause acute hypersensitivity reactions through mast cell activation, with a recent study attributing this effect to activation of the MAS-related G protein-coupled receptor X2 (MRGPRX2). In the present study, HEK293 cells expressing human MRGPRX2 and the human mast cell line LAD2 were used to characterize the activity of the broader family of polymyxins. Octapeptin C4, polymyxin B and colistin produced concentration-dependent calcium mobilization, degranulation, and CCL-2 (MCP-1) release in LAD2 mast cells, with the former being highly potent. CRISPR-Cas9 knockdown of MRGPRX2 in LAD2 cells and a MRGPRX2 inverse agonist caused a significant reduction in calcium mobilization, degranulation, and CCL-2 release, demonstrating dependency on MRGPRX2 expression. In contrast, polymyxin nonapeptides were far less potent calcium mobilisers and failed to induce functional degranulation in LAD2 cells. Our results confirm that activation of mast cells induced by polymyxin-related antibiotics is MRGPRX2-dependent and reveal that octapeptin C4 might be more liable, whilst nonapeptides are less liable, to trigger immediate hypersensitivity reactions clinically. The mechanism underpinning the difference in MRGPRX2 activation between polymyxin-related antibiotics is important to better understand as it may help design new, safer polymyxins and guide the optimal clinical use of existing polymyxin drugs.
Collapse
Affiliation(s)
- Jie Ding
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Stephanie S Zhang
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Nithya A Fernandopulle
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - John A Karas
- School of Chemistry, The University of Melbourne, Parkville, Victoria, Australia
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - James Ziogas
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Tony Velkov
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Graham A Mackay
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
3
|
Yoshino N, Yokoyama T, Sakai H, Sugiyama I, Odagiri T, Kimura M, Hojo W, Saino T, Muraki Y. Suitability of Polymyxin B as a Mucosal Adjuvant for Intranasal Influenza and COVID-19 Vaccines. Vaccines (Basel) 2023; 11:1727. [PMID: 38006059 PMCID: PMC10675063 DOI: 10.3390/vaccines11111727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Polymyxin B (PMB) is an antibiotic that exhibits mucosal adjuvanticity for ovalbumin (OVA), which enhances the immune response in the mucosal compartments of mice. Frequent breakthrough infections of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants indicate that the IgA antibody levels elicited by the mRNA vaccines in the mucosal tissues were insufficient for the prophylaxis of this infection. It remains unknown whether PMB exhibits mucosal adjuvanticity for antigens other than OVA. This study investigated the adjuvanticity of PMB for the virus proteins, hemagglutinin (HA) of influenza A virus, and the S1 subunit and S protein of SARS-CoV-2. BALB/c mice immunized either intranasally or subcutaneously with these antigens alone or in combination with PMB were examined, and the antigen-specific antibodies were quantified. PMB substantially increased the production of antigen-specific IgA antibodies in mucosal secretions and IgG antibodies in plasma, indicating its adjuvanticity for both HA and S proteins. This study also revealed that the PMB-virus antigen complex diameter is crucial for the induction of mucosal immunity. No detrimental effects were observed on the nasal mucosa or olfactory bulb. These findings highlight the potential of PMB as a safe candidate for intranasal vaccination to induce mucosal IgA antibodies for prophylaxis against mucosally transmitted infections.
Collapse
Affiliation(s)
- Naoto Yoshino
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba 028-3694, Iwate, Japan
| | - Takuya Yokoyama
- Department of Anatomy (Cell Biology), Iwate Medical University, 1-1-1 Idaidori, Yahaba 028-3694, Iwate, Japan
- Laboratory of Veterinary Anatomy and Cell Biology, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka 020-8550, Iwate, Japan
| | - Hironori Sakai
- R&D, Cellspect Co., Ltd., 2-4-23 Kitaiioka, Morioka 020-0857, Iwate, Japan
| | - Ikumi Sugiyama
- Division of Advanced Pharmaceutics, Department of Clinical Pharmaceutical Science, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba 028-3694, Iwate, Japan
| | - Takashi Odagiri
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba 028-3694, Iwate, Japan
| | - Masahiro Kimura
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba 028-3694, Iwate, Japan
| | - Wataru Hojo
- R&D, Cellspect Co., Ltd., 2-4-23 Kitaiioka, Morioka 020-0857, Iwate, Japan
| | - Tomoyuki Saino
- Department of Anatomy (Cell Biology), Iwate Medical University, 1-1-1 Idaidori, Yahaba 028-3694, Iwate, Japan
| | - Yasushi Muraki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba 028-3694, Iwate, Japan
| |
Collapse
|
4
|
Ontiveros-Padilla L, Batty CJ, Hendy DA, Pena ES, Roque JA, Stiepel RT, Carlock MA, Simpson SR, Ross TM, Abraham SN, Staats HF, Bachelder EM, Ainslie KM. Development of a broadly active influenza intranasal vaccine adjuvanted with self-assembled particles composed of mastoparan-7 and CpG. Front Immunol 2023; 14:1103765. [PMID: 37033992 PMCID: PMC10081679 DOI: 10.3389/fimmu.2023.1103765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
Currently licensed vaccine adjuvants offer limited mucosal immunity, which is needed to better combat respiratory infections such as influenza. Mast cells (MCs) are emerging as a target for a new class of mucosal vaccine adjuvants. Here, we developed and characterized a nanoparticulate adjuvant composed of an MC activator [mastoparan-7 (M7)] and a TLR ligand (CpG). This novel nanoparticle (NP) adjuvant was co-formulated with a computationally optimized broadly reactive antigen (COBRA) for hemagglutinin (HA), which is broadly reactive against influenza strains. M7 was combined at different ratios with CpG and tested for in vitro immune responses and cytotoxicity. We observed significantly higher cytokine production in dendritic cells and MCs with the lowest cytotoxicity at a charge-neutralizing ratio of nitrogen/phosphate = 1 for M7 and CpG. This combination formed spherical NPs approximately 200 nm in diameter with self-assembling capacity. Mice were vaccinated intranasally with COBRA HA and M7-CpG NPs in a prime-boost-boost schedule. Vaccinated mice had significantly higher antigen-specific antibody responses (IgG and IgA) in serum and mucosa compared with controls. Splenocytes from vaccinated mice had significantly increased cytokine production upon antigen recall and the presence of central and effector memory T cells in draining lymph nodes. Finally, co-immunization with NPs and COBRA HA induced influenza H3N2-specific HA inhibition antibody titers across multiple strains and partially protected mice from a challenge against an H3N2 virus. These results illustrate that the M7-CpG NP adjuvant combination can induce a protective immune response with a broadly reactive influenza antigen via mucosal vaccination.
Collapse
Affiliation(s)
- Luis Ontiveros-Padilla
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Cole J. Batty
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Dylan A. Hendy
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Erik S. Pena
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
| | - John A. Roque
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rebeca T. Stiepel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Michael A. Carlock
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Sean R. Simpson
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ted M. Ross
- Florida Research and Innovation Center, Port Saint, Cleveland Clinic Florida, Port St. Lucie, FL, United States
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Soman N. Abraham
- Departments of Pathology, Molecular Genetics and Microbiology and Immunology, Duke University School of Medicine, Durham, NC, United States
| | - Herman F. Staats
- Department of Pathology, School of Medicine, Duke University, Durham, NC, United States
- Duke Human Vaccines Institute, School of Medicine, Duke University, Durham, NC, United States
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Kristy M. Ainslie,
| |
Collapse
|
5
|
Kawamura H, Yoshino N, Murakami K, Kawamura H, Sugiyama I, Sasaki Y, Odagiri T, Sadzuka Y, Muraki Y. The relationship between the chemical structure, physicochemical properties, and mucosal adjuvanticity of sugar-based surfactants. Eur J Pharm Biopharm 2023; 182:1-11. [PMID: 36455784 DOI: 10.1016/j.ejpb.2022.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/07/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022]
Abstract
The relationship between the chemical structure, physicochemical properties, and mucosal adjuvanticity of sugar-based surfactants (SBSs) has not been sufficiently elucidated. Thus, in the present study, we systematically analyzed 11 SBSs for mucosal adjuvanticity. Ovalbumin (OVA)-specific antibody titers were measured in mice immunized intranasally with OVA plus SBS. We found that four SBSs (trehalose monododecanoate, sucrose monododecanoate, n-dodecyl-α-d-maltopyranoside, and n-dodecyl-β-d-maltopyranoside) exhibited the most potent adjuvanticity. We identified the following associations between chemical structure and adjuvanticity: 1) OVA-specific antibody titer increased with an increasing number of carbon atoms in the alkyl chain; 2) the adjuvanticity was not affected by the type of sugar or bond between the sugar and alkyl chain; and 3) SBSs with rigid structures exhibited less adjuvanticity. The relationship between physicochemical properties and adjuvanticity was as follows: 1) SBSs exhibited adjuvanticity above the critical micelle concentration and 2) in the SBSs with potent adjuvanticity, the diameter of the SBS-OVA complex was 70-75 nm. Our study indicates evidence for the direct involvement of chemical structure and physicochemical properties in determining adjuvanticity in SBSs.
Collapse
Affiliation(s)
- Hanae Kawamura
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Naoto Yoshino
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan.
| | - Kazuyuki Murakami
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Hideki Kawamura
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan; Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Ikumi Sugiyama
- Division of Advanced Pharmaceutics, Department of Clinical Pharmaceutical Sciences, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yutaka Sasaki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Takashi Odagiri
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yasuyuki Sadzuka
- Division of Advanced Pharmaceutics, Department of Clinical Pharmaceutical Sciences, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yasushi Muraki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| |
Collapse
|
6
|
Mardikasari SA, Sipos B, Csóka I, Katona G. Nasal route for antibiotics delivery: Advances, challenges and future opportunities applying the quality by design concepts. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
7
|
Luo J, Liu S, Lu H, Chen Q, Shi Y. A comprehensive review of microorganism-derived cyclic peptides: Bioactive functions and food safety applications. Compr Rev Food Sci Food Saf 2022; 21:5272-5290. [PMID: 36161470 DOI: 10.1111/1541-4337.13038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/09/2022] [Accepted: 08/21/2022] [Indexed: 01/28/2023]
Abstract
Cyclic peptides possess advanced structural characteristics of stability and play a vital role in medical treatment and agriculture. However, the biological functions of microorganism-derived cyclic peptides (MDCPs) and their applications in food industry were relatively absent. MDCPs are derived from extensive fermented food or soil. In this review, the synthesis approaches and structural characteristics are overviewed, while the interrelationship between bioactivities and functions is emphasized. This review summarizes the bioactivities of MDCPs from in vitro to in vivo, including antimicrobial activities, immune regulation, and antiviral cell activation. Their multiple functions as well as applications during food product processing, packaging, and storage are also comprehensively reviewed. Remarkably, some potential risks and cytotoxicity of MDCPs are also critically discussed. Moreover, future applications of MDCPs in the development of novel food additives and bioengineering materials are organized. Based on this review of native MDCPs, it is noteworthy that expected improvements of synthetic cyclic peptides in bioactive properties present potential valuable applications in future food, including artificial meat.
Collapse
Affiliation(s)
- Jiaqi Luo
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| | - Siyu Liu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| | - Hongyun Lu
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| | - Qihe Chen
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| | - Ying Shi
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Yoshino N, Kawamura H, Sugiyama I, Sasaki Y, Odagiri T, Sadzuka Y, Muraki Y. A systematic assessment of the relationship between synthetic surfactants and mucosal adjuvanticity. Eur J Pharm Biopharm 2021; 165:113-126. [PMID: 34004335 DOI: 10.1016/j.ejpb.2021.05.010] [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: 08/28/2020] [Revised: 03/24/2021] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
Intranasal immunization with surfactants as vaccine adjuvants enhances protective immunity against invasive mucosal pathogens. However, the effects of surfactants and their adjuvanticity on mucosal immune responses remain unclear. Comparison of the mucosal adjuvanticity of 20 water-soluble surfactants from the four classes based upon the polarity composition of the hydrophilic headgroup revealed that the order of mucosal adjuvanticity was as follows: amphoteric > nonionic > cationic > anionic. Within the same class, each surfactant displayed different adjuvanticity values. Analysis of the diameter and ζ-potential of amphoteric surfactant-OVA complexes and their surface physicochemical properties revealed that the diameter was approximately 100 nm, which is considered suitable for immune induction, and that the ζ-potential of the anionic surfactant-OVA complexes was exceedingly negative. The increase in the number of carbon atoms in the hydrophobic tailgroups of the amphoteric surfactant resulted in an increase in the OVA-specific Ab titers. Our findings demonstrate that amphoteric surfactants exhibit potent mucosal adjuvanticity and highlight the importance of the number of carbon atoms in the tailgroups and the diameter and ζ-potential of the complexes when designing mucosal adjuvants.
Collapse
Affiliation(s)
- Naoto Yoshino
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan.
| | - Hanae Kawamura
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, 2-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Ikumi Sugiyama
- Division of Advanced Pharmaceutics, Department of Clinical Pharmaceutical Sciences, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yutaka Sasaki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Takashi Odagiri
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yasuyuki Sadzuka
- Division of Advanced Pharmaceutics, Department of Clinical Pharmaceutical Sciences, School of Pharmacy, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Yasushi Muraki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, 1-1-1 Idaidori, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| |
Collapse
|
9
|
Willows S, Kulka M. Harnessing the Power of Mast Cells in unconventional Immunotherapy Strategies and Vaccine Adjuvants. Cells 2020; 9:cells9122713. [PMID: 33352850 PMCID: PMC7766453 DOI: 10.3390/cells9122713] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/03/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Mast cells are long-lived, granular, myeloid-derived leukocytes that have significant protective and repair functions in tissues. Mast cells sense disruptions in the local microenvironment and are first responders to physical, chemical and biological insults. When activated, mast cells release growth factors, proteases, chemotactic proteins and cytokines thereby mobilizing and amplifying the reactions of the innate and adaptive immune system. Mast cells are therefore significant regulators of homeostatic functions and may be essential in microenvironmental changes during pathogen invasion and disease. During infection by helminths, bacteria and viruses, mast cells release antimicrobial factors to facilitate pathogen expulsion and eradication. Mast cell-derived proteases and growth factors protect tissues from insect/snake bites and exposure to ultraviolet radiation. Finally, mast cells release mediators that promote wound healing in the inflammatory, proliferative and remodelling stages. Since mast cells have such a powerful repertoire of functions, targeting mast cells may be an effective new strategy for immunotherapy of disease and design of novel vaccine adjuvants. In this review, we will examine how certain strategies that specifically target and activate mast cells can be used to treat and resolve infections, augment vaccines and heal wounds. Although these strategies may be protective in certain circumstances, mast cells activation may be deleterious if not carefully controlled and any therapeutic strategy using mast cell activators must be carefully explored.
Collapse
Affiliation(s)
- Steven Willows
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Dr, Edmonton, AB T6G 2M9, Canada;
| | - Marianna Kulka
- Nanotechnology Research Centre, National Research Council Canada, 11421 Saskatchewan Dr, Edmonton, AB T6G 2M9, Canada;
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1, Canada
- Correspondence: ; Tel.: +1-780-641-1687
| |
Collapse
|
10
|
Mast Cell Functions Linking Innate Sensing to Adaptive Immunity. Cells 2020; 9:cells9122538. [PMID: 33255519 PMCID: PMC7761480 DOI: 10.3390/cells9122538] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
Although mast cells (MCs) are known as key drivers of type I allergic reactions, there is increasing evidence for their critical role in host defense. MCs not only play an important role in initiating innate immune responses, but also influence the onset, kinetics, and amplitude of the adaptive arm of immunity or fine-tune the mode of the adaptive reaction. Intriguingly, MCs have been shown to affect T-cell activation by direct interaction or indirectly, by modifying the properties of antigen-presenting cells, and can even modulate lymph node-borne adaptive responses remotely from the periphery. In this review, we provide a summary of recent findings that explain how MCs act as a link between the innate and adaptive immunity, all the way from sensing inflammatory insult to orchestrating the final outcome of the immune response.
Collapse
|
11
|
Alkanfari I, Freeman KB, Roy S, Jahan T, Scott RW, Ali H. Small-Molecule Host-Defense Peptide Mimetic Antibacterial and Antifungal Agents Activate Human and Mouse Mast Cells via Mas-Related GPCRs. Cells 2019; 8:cells8040311. [PMID: 30987258 PMCID: PMC6523814 DOI: 10.3390/cells8040311] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 12/18/2022] Open
Abstract
Host-defense peptides (HDPs) have an important therapeutic potential against microbial infections but their metabolic instability and cellular cytotoxicity have limited their utility. To overcome these limitations, we utilized five small-molecule, nonpeptide HDP mimetics (smHDPMs) and tested their effects on cytotoxicity, antimicrobial activity, and mast cell (MC) degranulation. None of the smHDPMs displayed cytotoxicity against mouse 3T3 fibroblasts or human transformed liver HepG2 cells. However, one compound had both antifungal and antibacterial activity. Surprisingly, all five compounds induced degranulation in a human MC line, LAD2, and this response was substantially reduced in Mas-related G protein-coupled receptor (GPCR)-X2 (MRGPRX2)-silenced cells. Furthermore, all five compounds induced degranulation in RBL-2H3 cells expressing MRGPRX2 but this response was abolished in cells expressing naturally occurring loss-of-function missense variants G165E (rs141744602) and D184H (rs372988289). Mrgprb2 is the likely mouse ortholog of human MRGPRX2, which is expressed in connective tissue MCs (CTMCs) such as cutaneous and peritoneal MCs (PMCs). All five smHDPMs induced degranulation in wild-type PMCs but not in cells derived from Mrgprb2−/− mice. These findings suggest that smHDPMs could serve as novel targets for the treatment of drug-resistant fungal and bacterial infections because of their ability to harness CTMCs’ host defense functions.
Collapse
Affiliation(s)
- Ibrahim Alkanfari
- Department of Pathology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA.
| | - Katie B Freeman
- Fox Chase Chemical Diversity Center, Doylestown, PA 18902, USA.
| | - Saptarshi Roy
- Department of Pathology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA.
| | - Tahsin Jahan
- Department of Pathology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA.
| | - Richard W Scott
- Fox Chase Chemical Diversity Center, Doylestown, PA 18902, USA.
| | - Hydar Ali
- Department of Pathology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
A Review of the Clinical Pharmacokinetics of Polymyxin B. Antibiotics (Basel) 2019; 8:antibiotics8010031. [PMID: 30909507 PMCID: PMC6466567 DOI: 10.3390/antibiotics8010031] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 03/12/2019] [Accepted: 03/15/2019] [Indexed: 11/29/2022] Open
Abstract
Polymyxin B remains an antibiotic of last resort because of its toxicities. Although newer therapies are becoming available, it is anticipated that resistance to these agents will continue to emerge, and understanding the safest and most efficacious manner to deliver polymyxin B will remain highly important. Recent data have demonstrated that polymyxin B may be less nephrotoxic than colistin. Pharmacokinetically, polymyxin B is primarily eliminated via non-renal pathways, and most do not recommend adjusting the dose for renal impairment. However, some recent studies suggest a weak relationship between polymyxin B clearance and patient creatinine clearance. This review article will describe the clinical pharmacokinetics of polymyxin B and address relevant issues in chemistry and assays available.
Collapse
|
13
|
Creighton RL, Woodrow KA. Microneedle-Mediated Vaccine Delivery to the Oral Mucosa. Adv Healthc Mater 2019; 8:e1801180. [PMID: 30537400 PMCID: PMC6476557 DOI: 10.1002/adhm.201801180] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/12/2018] [Indexed: 12/28/2022]
Abstract
The oral mucosa is a minimally invasive and immunologically rich site that is underutilized for vaccination due to physiological and immunological barriers. To develop effective oral mucosal vaccines, key questions regarding vaccine residence time, uptake, adjuvant formulation, dose, and delivery location must be answered. However, currently available dosage forms are insufficient to address all these questions. An ideal oral mucosal vaccine delivery system would improve both residence time and epithelial permeation while enabling efficient delivery of physicochemically diverse vaccine formulations. Microneedles have demonstrated these capabilities for dermal vaccine delivery. Additionally, microneedles enable precise control over delivery properties like depth, uniformity, and dosing, making them an ideal tool to study oral mucosal vaccination. Select studies have demonstrated the feasibility of microneedle-mediated oral mucosal vaccination, but they have only begun to explore the broad functionality of microneedles. This review describes the physiological and immunological challenges related to oral mucosal vaccine delivery and provides specific examples of how microneedles can be used to address these challenges. It summarizes and compares the few existing oral mucosal microneedle vaccine studies and offers a perspective for the future of the field.
Collapse
Affiliation(s)
- Rachel L Creighton
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
14
|
Ning Q. Main Complications of AECHB and Severe Hepatitis B (Liver Failure). ACUTE EXACERBATION OF CHRONIC HEPATITIS B 2019. [PMCID: PMC7498917 DOI: 10.1007/978-94-024-1603-9_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Qin Ning
- Department of Infectious Disease, Tongji Hospital, Wuhan, China
| |
Collapse
|
15
|
Arifuzzaman M, Mobley YR, Choi HW, Bist P, Salinas CA, Brown ZD, Chen SL, Staats HF, Abraham SN. MRGPR-mediated activation of local mast cells clears cutaneous bacterial infection and protects against reinfection. SCIENCE ADVANCES 2019; 5:eaav0216. [PMID: 30613778 PMCID: PMC6314830 DOI: 10.1126/sciadv.aav0216] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/27/2018] [Indexed: 05/22/2023]
Abstract
Mast cells (MCs) are strategically distributed at barrier sites and prestore various immunocyte-recruiting cytokines, making them ideal targets for selective activation to treat peripheral infections. Here, we report that topical treatment with mastoparan, a peptide MC activator (MCA), enhances clearance of Staphylococcus aureus from infected mouse skins and accelerates healing of dermonecrotic lesions. Mastoparan functions by activating connective tissue MCs (CTMCs) via the MRGPRX2 (Mas-related G protein-coupled receptor member X2) receptor. Peripheral CTMC activation, in turn, enhances recruitment of bacteria-clearing neutrophils and wound-healing CD301b+ dendritic cells. Consistent with MCs playing a master coordinating role, MC activation also augmented migration of various antigen-presenting dendritic cells to draining lymph nodes, leading to stronger protection against a second infection challenge. MCAs therefore orchestrate both the innate and adaptive immune arms, which could potentially be applied to combat peripheral infections by a broad range of pathogens.
Collapse
Affiliation(s)
- Mohammad Arifuzzaman
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Yuvon R. Mobley
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Hae Woong Choi
- Department of Pathology, Duke University, Durham, NC 27710, USA
| | - Pradeep Bist
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore
| | | | - Zachary D. Brown
- Undergraduate Program in Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Swaine L. Chen
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
- Infectious Diseases Group, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Herman F. Staats
- Department of Pathology, Duke University, Durham, NC 27710, USA
- Department of Immunology, Duke University, Durham, NC 27710, USA
- Department of Medicine, Duke University, Durham, NC 27710, USA
| | - Soman N. Abraham
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
- Department of Pathology, Duke University, Durham, NC 27710, USA
- Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore
- Department of Immunology, Duke University, Durham, NC 27710, USA
- Corresponding author.
| |
Collapse
|
16
|
Yoshino N, Takeshita R, Kawamura H, Murakami K, Sasaki Y, Sugiyama I, Sadzuka Y, Kagabu M, Sugiyama T, Muraki Y, Sato S. Critical micelle concentration and particle size determine adjuvanticity of cyclic lipopeptides. Scand J Immunol 2018; 88:e12698. [PMID: 29935085 DOI: 10.1111/sji.12698] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/01/2018] [Accepted: 06/20/2018] [Indexed: 12/20/2022]
Abstract
Cyclic lipopeptides such as surfactin and polymyxin have potent mucosal adjuvant properties. Cyclic lipopeptides are tensioactive compounds, but the relationship between adjuvanticity and surface activity is unknown. Here, we show that the critical micelle concentration (cmc) of surfactant and particle size of the surfactant-protein complex are important determinants of cyclic lipopeptide adjuvanticity. We found that the diameter of cyclic lipopeptide-ovalbumin (OVA) complex particles was significantly larger than that in the solutions of OVA alone at cyclic lipopeptide concentrations above the cmc. OVA-specific antibody titres in mice immunized intranasally with OVA and a cyclic lipopeptide at concentrations above its cmc were significantly higher than those in mice immunized with OVA plus the same dose of the cyclic lipopeptide but administered with formulations in which cyclic lipopeptide concentration was below the cmc. Thus, the concentration of the cyclic lipopeptide in the formulation at immunization, but not its overall dose, was critical for its adjuvanticity. Furthermore, two types of aggregates, the cyclic lipopeptide simplex micelles and the cyclic lipopeptide-OVA complex micelles, were found in formulations with SF concentrations above its cmc. Degranulation of mast cells exposed to SF simplex micelles was more pronounced when SF concentration was above the cmc. In conclusion, our study showed that surface activity properties, such as the cmc and the size of surfactant-protein complex, contribute to the adjuvanticity of cyclic lipopeptides. Our study proposes a novel idea that cmc is a key parameter for tensioactive adjuvants.
Collapse
Affiliation(s)
- Naoto Yoshino
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, Yahaba-cho, Japan
| | - Ryosuke Takeshita
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Hanae Kawamura
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Kazuyuki Murakami
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Yutaka Sasaki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, Yahaba-cho, Japan
| | - Ikumi Sugiyama
- Department of Advanced Pharmaceutics, School of Pharmacy, Iwate Medical University, Yahaba-cho, Japan
| | - Yasuyuki Sadzuka
- Department of Advanced Pharmaceutics, School of Pharmacy, Iwate Medical University, Yahaba-cho, Japan
| | - Masahiro Kagabu
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Toru Sugiyama
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Morioka, Japan
| | - Yasushi Muraki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, Yahaba-cho, Japan
| | - Shigehiro Sato
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, Yahaba-cho, Japan
| |
Collapse
|
17
|
Mast cell activators as novel immune regulators. Curr Opin Pharmacol 2018; 41:89-95. [PMID: 29843056 DOI: 10.1016/j.coph.2018.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/14/2018] [Indexed: 11/22/2022]
Abstract
Mast cells are an important cell type of the innate immune system that when activated, play a crucial role in generating protective innate host responses after bacterial and viral infection. Additionally, activated mast cells influence lymph node composition to regulate the induction of adaptive immune responses. The recognition that mast cells play a beneficial role in host responses to microbial infection and induction of adaptive immunity has provided the rationale to evaluate mast cell activators for use as antimicrobials or vaccine adjuvants. This review summarizes the role of mast cell activators in antimicrobial responses while also discussing the use of different classes of mast cell activators as potent vaccine adjuvants that enhance the induction of protective immune responses.
Collapse
|
18
|
Yoshino N, Takeshita R, Kawamura H, Sasaki Y, Kagabu M, Sugiyama T, Muraki Y, Sato S. Mast cells partially contribute to mucosal adjuvanticity of surfactin in mice. IMMUNITY INFLAMMATION AND DISEASE 2017; 6:117-127. [PMID: 29105371 PMCID: PMC5818442 DOI: 10.1002/iid3.204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/08/2017] [Accepted: 10/10/2017] [Indexed: 11/25/2022]
Abstract
Introduction Surfactin (SF) is a cyclic lipopeptide that has potent mucosal adjuvant properties. However, immunological mechanisms of SF adjuvant action have not yet been elucidated. As some cyclic lipopeptides, such as polymyxin, can stimulate histamine release from mast cells, we hypothesized that mast cell activation is critical for SF adjuvanticity. Methods/Results We observed that following intranasal immunization with ovalbumin (OVA) plus SF, the titers of the OVA‐specific antibody (Ab) in the mucosal secretions and plasma of mast cell‐deficient mice were significantly lower than those in congenic normal mice, although OVA‐specific Ab did not entirely disappear from mast cell‐deficient mice. SF induced degranulation of mast cells and release of histamine in vitro. To investigate whether SF stimulated mast cells in vivo, we measured body temperature of mice immunized intranasally with OVA plus SF because histamine level affects body temperature. Following immunizations, body temperature of immunized congenic normal mice transiently decreased, whereas body temperature of mast cell‐deficient mice did not change. Plasma levels of OVA‐specific IgE Ab were not significantly different in mast cell‐deficient and congenic normal mice. These findings suggest that SF directly affected mast cells in an IgE Ab‐independent fashion. Furthermore, we analyzed the effects of SF on MC/9 mast cells cultured in vitro. MC/9 cells stimulated by SF released not only histamine but also leukotriene B4 and prostaglandin D2. Moreover, SF up‐regulated mRNA expression levels of Tnf, Ccr5, and Il4 genes in mast cells. These cytokines may play a facilitating role in OVA‐specific immune responses in mice. Conclusion Overall, our results showed that mast cell activation partially mediated SF adjuvanticity.
Collapse
Affiliation(s)
- Naoto Yoshino
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Ryosuke Takeshita
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Hanae Kawamura
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Yutaka Sasaki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Masahiro Kagabu
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Toru Sugiyama
- Department of Obstetrics and Gynecology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Yasushi Muraki
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, Iwate, Japan
| | - Shigehiro Sato
- Division of Infectious Diseases and Immunology, Department of Microbiology, School of Medicine, Iwate Medical University, Iwate, Japan
| |
Collapse
|
19
|
Rhouma M, Beaudry F, Thériault W, Letellier A. Colistin in Pig Production: Chemistry, Mechanism of Antibacterial Action, Microbial Resistance Emergence, and One Health Perspectives. Front Microbiol 2016; 7:1789. [PMID: 27891118 PMCID: PMC5104958 DOI: 10.3389/fmicb.2016.01789] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/25/2016] [Indexed: 01/08/2023] Open
Abstract
Colistin (Polymyxin E) is one of the few cationic antimicrobial peptides commercialized in both human and veterinary medicine. For several years now, colistin has been considered the last line of defense against infections caused by multidrug-resistant Gram-negative such as Acinetobacter baumannii, Pseudomonas aeruginosa, and Klebsiella pneumoniae. Colistin has been extensively used orally since the 1960s in food animals and particularly in swine for the control of Enterobacteriaceae infections. However, with the recent discovery of plasmid-mediated colistin resistance encoded by the mcr-1 gene and the higher prevalence of samples harboring this gene in animal isolates compared to other origins, livestock has been singled out as the principal reservoir for colistin resistance amplification and spread. Co-localization of the mcr-1 gene and Extended-Spectrum-β-Lactamase genes on a unique plasmid has been also identified in many isolates from animal origin. The use of colistin in pigs as a growth promoter and for prophylaxis purposes should be banned, and the implantation of sustainable measures in pig farms for microbial infection prevention should be actively encouraged and financed. The scientific research should be encouraged in swine medicine to generate data helping to reduce the exacerbation of colistin resistance in pigs and in manure. The establishment of guidelines ensuring a judicious therapeutic use of colistin in pigs, in countries where this drug is approved, is of crucial importance. The implementation of a microbiological withdrawal period that could reduce the potential contamination of consumers with colistin resistant bacteria of porcine origin should be encouraged. Moreover, the management of colistin resistance at the human-pig-environment interface requires the urgent use of the One Health approach for effective control and prevention. This approach needs the collaborative effort of multiple disciplines and close cooperation between physicians, veterinarians, and other scientific health and environmental professionals. This review is an update on the chemistry of colistin, its applications and antibacterial mechanism of action, and on Enterobacteriaceae resistance to colistin in pigs. We also detail and discuss the One Health approach and propose guidelines for colistin resistance management.
Collapse
Affiliation(s)
- Mohamed Rhouma
- Chaire de Recherche Industrielle du CRSNG en Salubrité des Viandes, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
- Groupe de Recherche et d’Enseignement en Salubrité Alimentaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
| | - Francis Beaudry
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
- Groupe de Recherche en Pharmacologie Animale du Québec, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
| | - William Thériault
- Chaire de Recherche Industrielle du CRSNG en Salubrité des Viandes, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
- Groupe de Recherche et d’Enseignement en Salubrité Alimentaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
| | - Ann Letellier
- Chaire de Recherche Industrielle du CRSNG en Salubrité des Viandes, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
- Groupe de Recherche et d’Enseignement en Salubrité Alimentaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
- Groupe de Recherche en Pharmacologie Animale du Québec, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-HyacintheQC, Canada
| |
Collapse
|
20
|
Kicielińska J, Szczygieł A, Rossowska J, Anger N, Kempińska K, Świtalska M, Kaszowska M, Wietrzyk J, Boratyński J, Pajtasz-Piasecka E. Oral Administration of Polymyxin B Modulates the Activity of Lipooligosaccharide E. coli B against Lung Metastases in Murine Tumor Models. PLoS One 2016; 11:e0148156. [PMID: 26829479 PMCID: PMC4735115 DOI: 10.1371/journal.pone.0148156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 01/13/2016] [Indexed: 01/18/2023] Open
Abstract
Introduction Polymyxin B (PmB) belongs to the group of cyclic peptide antibiotics, which neutralize the activity of LPS by binding to lipid A. The aim of this study was to analyze the effect of PmB on the biological activity of lipooligosaccharide (LOS E. coli B,rough form of LPS) in vitro and in experimental metastasis models. Results Cultures of murine macrophage J774A.1 cells and murine bone marrow-derived dendritic cells (BM-DC) stimulated in vitro with LOS and supplemented with PmB demonstrated a decrease in inflammatory cytokine production (IL-6, IL-10, TNF-α) and down-regulation of CD40, CD80, CD86 and MHC class II molecule expression. Additionally, PmB suspended in drinking water was given to the C57BL/6 mice seven or five days prior to the intravenous injection of B16 or LLC cells and intraperitoneal application of LOS. This strategy of PmB administration was continued throughout the duration of the experiments (29 or 21 days). In B16 model, statistically significant decrease in the number of metastases in mice treated with PmB and LOS (p<0.01) was found on the 14th day of the experiments, whereas the most intensive changes in surface-antigen expression and ex vivo production of IL-6, IL-1β and TNF-α by peritoneal cells were observed 7 days earlier. By contrast, antigen expression and ex vivo production of IL-6, IL-10, IFN-γ by splenocytes remained relatively high and stable. Statistically significant decrease in LLC metastases number was observed after the application of LOS (p<0.01) and in the group of mice preconditioned by PmB and subsequently treated with LOS (LOS + PmB, p<0.01). Conclusions In conclusion, prolonged in vivo application of PmB was not able to neutralize the LOS-induced immune cell activity but its presence in the organism of treated mice was important in modulation of the LOS-mediated response against the development of metastases.
Collapse
Affiliation(s)
- Jagoda Kicielińska
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Agnieszka Szczygieł
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Joanna Rossowska
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Natalia Anger
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Katarzyna Kempińska
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Marta Świtalska
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Marta Kaszowska
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Joanna Wietrzyk
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Janusz Boratyński
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Elżbieta Pajtasz-Piasecka
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
- * E-mail:
| |
Collapse
|
21
|
Nizard M, Diniz MO, Roussel H, Tran T, Ferreira LC, Badoual C, Tartour E. Mucosal vaccines: novel strategies and applications for the control of pathogens and tumors at mucosal sites. Hum Vaccin Immunother 2015; 10:2175-87. [PMID: 25424921 DOI: 10.4161/hv.29269] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mucosal immune system displays several adaptations reflecting the exposure to the external environment. The efficient induction of mucosal immune responses also requires specific approaches, such as the use of appropriate administration routes and specific adjuvants and/or delivery systems. In contrast to vaccines delivered via parenteral routes, experimental, and clinical evidences demonstrated that mucosal vaccines can efficiently induce local immune responses to pathogens or tumors located at mucosal sites as well as systemic response. At least in part, such features can be explained by the compartmentalization of mucosal B and T cell populations that play important roles in the modulation of local immune responses. In the present review, we discuss molecular and cellular features of the mucosal immune system as well as novel immunization approaches that may lead to the development of innovative and efficient vaccines targeting pathogens and tumors at different mucosal sites.
Collapse
Affiliation(s)
- Mevyn Nizard
- a INSERM U970; Universite Paris Descartes; Sorbonne Paris-Cité; Paris, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Fang Y, Xiang Z. Roles and relevance of mast cells in infection and vaccination. J Biomed Res 2015; 30:253-63. [PMID: 26565602 PMCID: PMC4946316 DOI: 10.7555/jbr.30.20150038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 04/08/2015] [Accepted: 04/26/2015] [Indexed: 01/06/2023] Open
Abstract
In addition to their well-established role in allergy mast cells have been described as contributing to functional regulation of both innate and adaptive immune responses in host defense. Mast cells are of hematopoietic origin but typically complete their differentiation in tissues where they express immune regulatory functions by releasing diverse mediators and cytokines. Mast cells are abundant at mucosal tissues which are portals of entry for common infectious agents in addition to allergens. Here, we review the current understanding of the participation of mast cells in defense against infection. We also discuss possibilities of exploiting mast cell activation to provide adequate adjuvant activity that is needed in high-quality vaccination against infectious diseases.
Collapse
Affiliation(s)
- Yu Fang
- Department of Microbiology and Immunology; Clinical Research Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550025, China
| | - Zou Xiang
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, University of Gothenburg, Gothenburg 40530, Sweden.
| |
Collapse
|
23
|
Patel S, Ahmed S, Eswari JS. Therapeutic cyclic lipopeptides mining from microbes: latest strides and hurdles. World J Microbiol Biotechnol 2015; 31:1177-93. [PMID: 26041368 DOI: 10.1007/s11274-015-1880-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 05/31/2015] [Indexed: 12/23/2022]
Abstract
Infectious diseases impose serious public health burdens and often have devastating consequences. The cyclic lipopeptides elaborated by bacteria Bacillus, Paenibacillus, Pseudomonas, Streptomyces, Serratia, Propionibacterium and fungus Fusarium are very crucial in restraining the pathogens. Composed of a peptide and a fatty acyl moiety these amphiphilic metabolites exhibit broad spectrum antimicrobial effects. Among the plethora of cyclic lipopeptides, only selective few have emerged as robust antibiotics. For their functional vigor, polymyxin, daptomycin, surfactin, iturin, fengysin, paenibacterin and pseudofactin have been integrated in mainstream healthcare. Daptomycin has been a significant part of antimicrobial arsenal since the past decade. As the magnitude of drug resistance rises in unprecedented manner, the urgency of prospecting novel cyclic lipopeptides is being perceived. Intense research has revealed the implication of these bioactive compounds stretching beyond antibacterial and antifungal. Anticancer, immunomodulatory, prosthetic parts disinfection and vaccine adjuvancy are some of the validated prospects. This review discusses the emerging applications, mechanisms governing the biological actions, role of genomics in refining structure and function, semi-synthetic analog discovery, novel strain isolation, setbacks etc. Though its beyond the scope of the current topic, for holistic purpose, the role of lipopeptides in bioremediation and crop biotechnology has been briefly outlined. This updated critique is expected to galvanize innovations and diversify therapeutic recruitment of microbial lipopeptides.
Collapse
Affiliation(s)
- Seema Patel
- Bioinformatics and Medical Informatics Research Center, San Diego State University, San Diego, CA, 92182, USA,
| | | | | |
Collapse
|
24
|
Bento D, Staats HF, Gonçalves T, Borges O. Development of a novel adjuvanted nasal vaccine: C48/80 associated with chitosan nanoparticles as a path to enhance mucosal immunity. Eur J Pharm Biopharm 2015; 93:149-64. [PMID: 25818119 DOI: 10.1016/j.ejpb.2015.03.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 02/19/2015] [Accepted: 03/20/2015] [Indexed: 11/15/2022]
Abstract
In a time in which mucosal vaccines development has been delayed by the lack of safe and effective mucosal adjuvants, the combination of adjuvants has started to be explored as a strategy to obtain potent vaccine formulations. This study describes a novel adjuvant combination as an effective approach for a nasal vaccine - the association of the mast cell activator compound 48/80 with chitosan based nanoparticles. It was hypothesized that mucoadhesive nanoparticles would promote the cellular uptake and prolong the antigen residence time on nasal cavity. Simultaneously, mast cell activation would promote a local microenvironment favorable to the development of an immune response. To test this hypothesis, two different C48/80 loaded nanoparticles (NPs) were prepared: Chitosan-C48/80 NP (Chi-C48/80 NP) and Chitosan/Alginate-C48/80 NP (Chi/Alg-C48/80 NP). The potential as a vaccine adjuvant of the two delivery systems was evaluated and directly compared. Both formulations had a mean size near 500nm and a positive charge; however, Chi-C48/80 NP was a more effective adjuvant delivery system when compared with Chi/Alg-C48/80 NP or C48/80 alone. Chi-C48/80 NP activated mast cells at a greater extent, were better internalized by antigen presenting cells than Chi/Alg-C48/80 NP and successfully enhanced the nasal residence time of a model antigen. Superiority of Chi-C48/80 NP as adjuvant was also observed in vivo. Therefore, nasal immunization of mice with Bacillus anthracis protective antigen (PA) adsorbed on Chi-C48/80 NP elicited high levels of serum anti-PA neutralizing antibodies and a more balanced Th1/Th2 profile than C48/80 in solution or Chi/Alg-C48/80 NP. The incorporation of C48/80 within Chi NP also promoted a mucosal immunity greater than all the other adjuvanted groups tested, showing that the combination of a mast cell activator and chitosan NP could be a promising strategy for nasal immunization.
Collapse
Affiliation(s)
- D Bento
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - H F Staats
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - T Gonçalves
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Institute of Microbiology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - O Borges
- Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|