1
|
Oladipo EK, Adeyemo SF, Oshoneye AI, Akintola HB, Elegbede BI, Ayoomoba TU, Atilade DA, Adegboye OO, Ejikeme AE, Balogun CO, Aderibigbe KA, Popoola PO, Alabi VA, Irewolede BA, Ano-Edward GH, Ayeleso AO, Onyeaka H. Harnessing computational immunology to design targeted subunit vaccines for infectious bursal disease in poultry. FRONTIERS IN BIOINFORMATICS 2025; 5:1562997. [PMID: 40255694 PMCID: PMC12006097 DOI: 10.3389/fbinf.2025.1562997] [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: 01/21/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction Infectious bursal disease (IBD), caused by the infectious bursal disease Q8 virus (IBDV), is a highly contagious disease in young chickens, leading to immunosuppression with great economic importance. IBDV, a non-enveloped virus with a bipartite dsRNA genome, infects the bursa of Fabricius, causing severe gastrointestinal disease. Effective vaccines are urgently needed due to the limitations of current oral vaccines, including gastrointestinal degradation and low immunogenicity. This study designs and evaluates a multiepitope subunit vaccine using immunoinformatics. Methods Sequences of the IBDV structural proteins VP2 and VP3 were obtained from the National Centre for Biotechnology Information) NCBI. These are structural proteins VP2 and VP3 were subjected to the Vaxijen 2.0 webserver to predict the antigenicity, ToxiPred to predict the toxicity and further analyzed to identify immunogenic epitopes of Chicken Leukocyte Antigens (CLAs) using the NetMHCpan 4.1 webserver. Results The final vaccine construct includes 2 HTL, 21 CTL, and 7 LBL epitopes, with gallinacin-3 precursor as an adjuvant. The construct is antigenic (0.5605), non-allergenic, and non-toxic, consisting of 494 amino acids with a molecular weight of 54.88 kDa and a positive charge (pI of 9.23). It is stable, hydrophilic, and soluble. Population coverage analysis revealed a global immune coverage of 89.83%, with the highest in Europe (99.86%) and the lowest in Central America (25.01%). Molecular docking revealed strong interactions with TLR-2_1, TLR-4, and TLR-7, with TLR-7 exhibiting the highest binding affinity (-366.15 kcal/mol). Immune simulations indicated a robust immune response, with high initial IgM levels, sustained IgG, memory cell formation, and activation of T helper (Th) cells 1 and 2, Natural Killer (NK) cells, and dendritic cells, suggesting potential long-lasting immunity against IBDV. Discussion This study presents a promising multi-epitope subunit vaccine candidate capable of effective immunization against IBDV with broad population coverage. However, further in vivo experimental validation is required to confirm its efficacy and safety.
Collapse
Affiliation(s)
- Elijah Kolawole Oladipo
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
- Department of Microbiology, Laboratory of Molecular Biology, Immunology and Bioinformatics, Adeleke University, Ede, Osun, Nigeria
- Department of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| | - Stephen Feranmi Adeyemo
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Ayomiposi Isaiah Oshoneye
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Hannah Blessing Akintola
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Bolatito Islam Elegbede
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Tobiloba Uren Ayoomoba
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Dorcas Ayomide Atilade
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Omolara Omoboye Adegboye
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Abuoma Elizabeth Ejikeme
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
- Department of Biomedical Laboratory Science, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Chris Olamide Balogun
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Kehinde Abolade Aderibigbe
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Possible Okikiola Popoola
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Victoria Ajike Alabi
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | - Boluwatife Ayobami Irewolede
- Division of Vaccine and Pharmacotherapies Design and Development, Helix Biogen Institute, Ogbomoso, Oyo, Nigeria
| | | | - Ademola Olabode Ayeleso
- Department of Biochemistry, Bowen University, Iwo, Osun, Nigeria
- Department of Life and Consumer Sciences, University of South Africa, Florida Park, Roodeport, South Africa
| | - Helen Onyeaka
- Department of Chemical Engineering, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
2
|
Berber E, Mulik S, Rouse BT. Meeting the Challenge of Controlling Viral Immunopathology. Int J Mol Sci 2024; 25:3935. [PMID: 38612744 PMCID: PMC11011832 DOI: 10.3390/ijms25073935] [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: 02/14/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
The mission of this review is to identify immune-damaging participants involved in antiviral immunoinflammatory lesions. We argue these could be targeted and their activity changed selectively by maneuvers that, at the same time, may not diminish the impact of components that help resolve lesions. Ideally, we need to identify therapeutic approaches that can reverse ongoing lesions that lack unwanted side effects and are affordable to use. By understanding the delicate balance between immune responses that cause tissue damage and those that aid in resolution, novel strategies can be developed to target detrimental immune components while preserving the beneficial ones. Some strategies involve rebalancing the participation of immune components using various approaches, such as removing or blocking proinflammatory T cell products, expanding regulatory cells, restoring lost protective cell function, using monoclonal antibodies (moAb) to counteract inhibitory molecules, and exploiting metabolic differences between inflammatory and immuno-protective responses. These strategies can help reverse ongoing viral infections. We explain various approaches, from model studies and some clinical evidence, that achieve innate and adaptive immune rebalancing, offering insights into potential applications for controlling chronic viral-induced lesions.
Collapse
Affiliation(s)
- Engin Berber
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Sachin Mulik
- Center for Biomedical Research, The University of Texas Health Science Center at Tyler, Tyler, TX 75708, USA;
| | - Barry T. Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
3
|
Yu YB, Liu Y, Li S, Yang XY, Guo Z. The pH-responsive zeolitic imidazolate framework nanoparticle as a promising immune-enhancing adjuvant for anti-caries vaccine. J Dent 2023; 130:104413. [PMID: 36634754 DOI: 10.1016/j.jdent.2023.104413] [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: 07/28/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES Streptococcus mutans (S. mutans) is the main aetiologic bacterium of dental caries, whose protein antigen (PAc) has been administered as an anti-caries vaccine. In addition, several fusion proteins or PAc combined with adjuvants were used as anti-caries vaccines to improve the relatively weak immunogenicity of PAc. However, there are no nanoparticle-based adjuvants with good biocompatibility, excellent biodegradability, or the high loading performance of antigens used for anti-caries vaccines. This study aimed to prepare an innovative nanoparticle-based anti-caries vaccine and evaluate immune responses elicited by this vaccine in vitro and in vivo. METHODS In this study, an anti-caries vaccine was prepared by an antigen of recombinant protein PAc from S. mutans and an adjuvant of zeolitic imidazolate framework-8 nanoparticles (ZIF-8 NPs) synthesized using a hydrothermal method. Then, mice were administrated intranasally by ZIF-8@PAc vaccine, and immune responses were evaluated. RESULTS ZIF-8 NPs not only greatly improved the internalization of the antigen but also released the PAc protein after degradation of ZIF-8 NPs in lysosomes for the further processing and presentation of antigen-presenting cells. In addition, ZIF-8@PAc induced significantly more potent PAc-specific serum IgG and saliva IgA antibodies, a higher splenocyte proliferation index, higher levels of the cytokines IL-4, IL-6, IL-10, IL-17A and IFN-γ, and a higher percentage of mature DCs and CD4+ memory T cells in vivo. CONCLUSIONS The ZIF-8 NPs, as an anti-caries vaccine adjuvant-assisted antigen PAc, elicit significantly potent immune responses, aiding in the further prevention of dental caries. CLINICAL SIGNIFICANCE Vaccine immunotherapy is an attractive strategy for prevention and treatment of dental caries. The ZIF-8@PAc vaccine can induce significantly high level of immune responses in this study, which indicates great potential for prevention and treatment of caries.
Collapse
Affiliation(s)
- You-Bo Yu
- Center for Biological Science and Technology, Guangdong Zhuhai-Macao Joint Biotech Laboratory, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China; Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Ying Liu
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Sha Li
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| | - Xiao-Yan Yang
- Zhuhai Key Laboratory of Basic and Applied Research in Chinese Medicine, College of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China.
| | - Zhong Guo
- Center for Biological Science and Technology, Guangdong Zhuhai-Macao Joint Biotech Laboratory, Advanced Institute of Natural Sciences, Beijing Normal University, Zhuhai, China; Key Laboratory of Cell Proliferation and Regulation Biology of Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China.
| |
Collapse
|
4
|
James J, Meyer SM, Hong HA, Dang C, Linh HTY, Ferreira W, Katsande PM, Vo L, Hynes D, Love W, Banyard AC, Cutting SM. Intranasal Treatment of Ferrets with Inert Bacterial Spores Reduces Disease Caused by a Challenging H7N9 Avian Influenza Virus. Vaccines (Basel) 2022; 10:vaccines10091559. [PMID: 36146637 PMCID: PMC9502451 DOI: 10.3390/vaccines10091559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Influenza is a respiratory infection that continues to present a major threat to human health, with ~500,000 deaths/year. Continued circulation of epidemic subtypes in humans and animals potentially increases the risk of future pandemics. Vaccination has failed to halt the evolution of this virus and next-generation prophylactic approaches are under development. Naked, “heat inactivated”, or inert bacterial spores have been shown to protect against influenza in murine models. Methods: Ferrets were administered intranasal doses of inert bacterial spores (DSM 32444K) every 7 days for 4 weeks. Seven days after the last dose, the animals were challenged with avian H7N9 influenza A virus. Clinical signs of infection and viral shedding were monitored. Results: Clinical symptoms of infection were significantly reduced in animals dosed with DSM 32444K. The temporal kinetics of viral shedding was reduced but not prevented. Conclusion: Taken together, nasal dosing using heat-stable spores could provide a useful approach for influenza prophylaxis in both humans and animals.
Collapse
Affiliation(s)
- Joe James
- Animal and Plant Health Agency (APHA), Woodham Lane, Weybridge KT15 3NB, Surrey, UK
| | - Stephanie M. Meyer
- Animal and Plant Health Agency (APHA), Woodham Lane, Weybridge KT15 3NB, Surrey, UK
| | - Huynh A. Hong
- SporeGen Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
| | - Chau Dang
- SporeGen Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
| | - Ho T. Y. Linh
- HURO Biotech JSC, Lot A1-8, VL3 Road, Vinh Loc 2 Industrial Park, Long Hiep Commune, Ben Luc District, Long An, Vietnam
| | - William Ferreira
- SporeGen Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
| | - Paidamoyo M. Katsande
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, Surrey, UK
| | - Linh Vo
- SporeGen Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
| | - Daniel Hynes
- Destiny Pharma Plc., Sussex Innovation Centre, Science Park Square, Falmer, Brighton BN1 9SB, UK
| | - William Love
- Destiny Pharma Plc., Sussex Innovation Centre, Science Park Square, Falmer, Brighton BN1 9SB, UK
| | - Ashley C. Banyard
- Animal and Plant Health Agency (APHA), Woodham Lane, Weybridge KT15 3NB, Surrey, UK
| | - Simon M. Cutting
- SporeGen Ltd., London Bioscience Innovation Centre, 2 Royal College Street, London NW1 0NH, UK
- Department of Biological Sciences, Royal Holloway University of London, Egham TW20 0EX, Surrey, UK
- Correspondence: ; Tel.: +44-(0)7900-408043
| |
Collapse
|
5
|
Ravichandran S, Manickam N, Kandasamy M. Liposome encapsulated clodronate mediated elimination of pathogenic macrophages and microglia: A promising pharmacological regime to defuse cytokine storm in COVID-19. MEDICINE IN DRUG DISCOVERY 2022; 15:100136. [PMID: 35721801 PMCID: PMC9190184 DOI: 10.1016/j.medidd.2022.100136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/14/2022] Open
Abstract
The emergence of new SARS-CoV-2 variants continues to pose an enormous public health concern. The SARS-CoV-2 infection disrupted host immune response accounting for cytokine storm has been linked to multiorgan failure and mortality in a significant portion of positive cases. Abruptly activated macrophages have been identified as the key pathogenic determinant of cytokine storm in COVID-19. Besides, reactive microglia have been known to discharge a surplus amount of proinflammatory factors leading to neuropathogenic events in the brains of SARS-CoV-2 infected individuals. Considering the fact, depletion of activated macrophages and microglia could be proposed to eradicate the life-threatening cytokine storm in COVID-19. Clodronate, a non-nitrogenous bisphosphonate drug has been identified as a potent macrophage and microglial depleting agent. While recent advancement in the field of liposome encapsulation technology offers the most promising biological tool for drug delivery, liposome encapsulated clodronate has been reported to effectively target and induce prominent phagocytic cell death in activated macrophages and microglia compared to free clodronate molecules. Thus, in this review article, we emphasize that depletion of activated macrophages and microglial cells by administration of liposome encapsulated clodronate can be a potential therapeutic strategy to diminish the pathogenic cytokine storm and alleviate multiorgan failure in COVID-19. Moreover, recently developed COVID-19 vaccines appear to render the chronic activation of macrophages accounting for immunological dysregulation in some cases. Therefore, the use of liposome encapsulated clodronate can also be extended to the clinical management of unforeseen immunogenic reactions resulting from activated macrophages associated adverse effects of COVID-19 vaccines.
Collapse
Affiliation(s)
- Sowbarnika Ravichandran
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Nivethitha Manickam
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Mahesh Kandasamy
- Laboratory of Stem Cells and Neuroregeneration, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
- University Grants Commission, Faculty Recharge Programme (UGC-FRP), New Delhi 110002, India
| |
Collapse
|
6
|
Girkin JLN, Maltby S, Bartlett NW. Toll-like receptor-agonist-based therapies for respiratory viral diseases: thinking outside the cell. Eur Respir Rev 2022; 31:210274. [PMID: 35508333 PMCID: PMC9488969 DOI: 10.1183/16000617.0274-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 11/24/2022] Open
Abstract
Respiratory virus infections initiate in the upper respiratory tract (URT). Innate immunity is critical for initial control of infection at this site, particularly in the absence of mucosal virus-neutralising antibodies. If the innate immune response is inadequate, infection can spread to the lower respiratory tract (LRT) causing community-acquired pneumonia (as exemplified by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)/coronavirus disease 2019). Vaccines for respiratory viruses (influenza and SARS-CoV-2) leverage systemic adaptive immunity to protect from severe lung disease. However, the URT remains vulnerable to infection, enabling viral transmission and posing an ongoing risk of severe disease in populations that lack effective adaptive immunity.Innate immunity is triggered by host cell recognition of viral pathogen-associated molecular patterns via molecular sensors such as Toll-like receptors (TLRs). Here we review the role of TLRs in respiratory viral infections and the potential of TLR-targeted treatments to enhance airway antiviral immunity to limit progression to severe LRT disease and reduce person-to-person viral transmission. By considering cellular localisation and antiviral mechanisms of action and treatment route/timing, we propose that cell surface TLR agonist therapies are a viable strategy for preventing respiratory viral diseases by providing immediate, durable pan-viral protection within the URT.
Collapse
Affiliation(s)
- Jason L N Girkin
- Viral Immunology and Respiratory Disease Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Steven Maltby
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Nathan W Bartlett
- Viral Immunology and Respiratory Disease Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| |
Collapse
|
7
|
Cui H, Zhang C, Zhang C, Cai Z, Chen L, Chen Z, Zhao K, Qiao S, Wang Y, Meng L, Dong S, Liu J, Guo Z. Anti-Influenza Effect and Mechanisms of Lentinan in an ICR Mouse Model. Front Cell Infect Microbiol 2022; 12:892864. [PMID: 35669119 PMCID: PMC9163413 DOI: 10.3389/fcimb.2022.892864] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/27/2022] [Indexed: 12/12/2022] Open
Abstract
Influenza virus is a serious threat to global human health and public health security. There is an urgent need to develop new anti-influenza drugs. Lentinan (LNT) has attracted increasing attention in recent years. As potential protective agent, LNT has been shown to have anti-tumor, anti-inflammatory, and antiviral properties. However, there has been no further research into the anti-influenza action of lentinan in vivo, and the mechanism is still not fully understood. In this study, the anti-influenza effect and mechanism of Lentinan were studied in the Institute of Cancer Research (ICR) mouse model. The results showed that Lentinan had a high degree of protection in mice against infection with influenza A virus, delayed the emergence of clinical manifestations, improved the survival rate of mice, significantly prolonged the middle survival days, attenuated the weight loss, and reduced the lung coefficient of mice. It alleviated the pathological damage of mice infected with the influenza virus and improved blood indices. Lentinan treatment considerably inhibited inflammatory cytokine (TNF-α, IL-1β, IL-4, IL-5, IL-6) levels in the serum and lung and improved IFN-γ cytokine levels, which reduced cytokine storms caused by influenza virus infection. The underlying mechanisms of action involved Lentinan inhibiting the inflammatory response by regulating the TLR4/MyD88 signaling pathway. This study provides a foundation for the clinical application of Lentinan, and provides new insight into the development of novel immunomodulators.
Collapse
Affiliation(s)
- Huan Cui
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Animal Medicine, Jilin University, Changchun, China
| | - Cheng Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Chunmao Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Zhuming Cai
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Ligong Chen
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Zhaoliang Chen
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Kui Zhao
- College of Animal Medicine, Jilin University, Changchun, China
| | - Sina Qiao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yingchun Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Lijia Meng
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Shishan Dong
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
- *Correspondence: Shishan Dong, ; Juxiang Liu, ; Zhendong Guo,
| | - Juxiang Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
- *Correspondence: Shishan Dong, ; Juxiang Liu, ; Zhendong Guo,
| | - Zhendong Guo
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- *Correspondence: Shishan Dong, ; Juxiang Liu, ; Zhendong Guo,
| |
Collapse
|
8
|
Shirey KA, Blanco JCG, Vogel SN. Targeting TLR4 Signaling to Blunt Viral-Mediated Acute Lung Injury. Front Immunol 2021; 12:705080. [PMID: 34282358 PMCID: PMC8285366 DOI: 10.3389/fimmu.2021.705080] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/10/2021] [Indexed: 01/14/2023] Open
Abstract
Respiratory viral infections have been a long-standing global burden ranging from seasonal recurrences to the unexpected pandemics. The yearly hospitalizations from seasonal viruses such as influenza can fluctuate greatly depending on the circulating strain(s) and the congruency with the predicted strains used for the yearly vaccine formulation, which often are not predicted accurately. While antiviral agents are available against influenza, efficacy is limited due to a temporal disconnect between the time of infection and symptom development and viral resistance. Uncontrolled, influenza infections can lead to a severe inflammatory response initiated by pathogen-associated molecular patterns (PAMPs) or host-derived danger-associated molecular patterns (DAMPs) that ultimately signal through pattern recognition receptors (PRRs). Overall, these pathogen-host interactions result in a local cytokine storm leading to acute lung injury (ALI) or the more severe acute respiratory distress syndrome (ARDS) with concomitant systemic involvement and more severe, life threatening consequences. In addition to traditional antiviral treatments, blocking the host's innate immune response may provide a more viable approach to combat these infectious pathogens. The SARS-CoV-2 pandemic illustrates a critical need for novel treatments to counteract the ALI and ARDS that has caused the deaths of millions worldwide. This review will examine how antagonizing TLR4 signaling has been effective experimentally in ameliorating ALI and lethal infection in challenge models triggered not only by influenza, but also by other ALI-inducing viruses.
Collapse
Affiliation(s)
- Kari Ann Shirey
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, United States
| | | | - Stefanie N. Vogel
- Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
9
|
Owen AM, Fults JB, Patil NK, Hernandez A, Bohannon JK. TLR Agonists as Mediators of Trained Immunity: Mechanistic Insight and Immunotherapeutic Potential to Combat Infection. Front Immunol 2021; 11:622614. [PMID: 33679711 PMCID: PMC7930332 DOI: 10.3389/fimmu.2020.622614] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/24/2020] [Indexed: 12/18/2022] Open
Abstract
Despite advances in critical care medicine, infection remains a significant problem that continues to be complicated with the challenge of antibiotic resistance. Immunocompromised patients are highly susceptible to development of severe infection which often progresses to the life-threatening condition of sepsis. Thus, immunotherapies aimed at boosting host immune defenses are highly attractive strategies to ward off infection and protect patients. Recently there has been mounting evidence that activation of the innate immune system can confer long-term functional reprogramming whereby innate leukocytes mount more robust responses upon secondary exposure to a pathogen for more efficient clearance and host protection, termed trained immunity. Toll-like receptor (TLR) agonists are a class of agents which have been shown to trigger the phenomenon of trained immunity through metabolic reprogramming and epigenetic modifications which drive profound augmentation of antimicrobial functions. Immunomodulatory TLR agonists are also highly beneficial as vaccine adjuvants. This review provides an overview on TLR signaling and our current understanding of TLR agonists which show promise as immunotherapeutic agents for combating infection. A brief discussion on our current understanding of underlying mechanisms is also provided. Although an evolving field, TLR agonists hold strong therapeutic potential as immunomodulators and merit further investigation for clinical translation.
Collapse
Affiliation(s)
- Allison M Owen
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jessica B Fults
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States.,University of Texas Southwestern Medical School, Dallas, TX, United States
| | - Naeem K Patil
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
10
|
Hasanzadeh S, Habibi M, Shokrgozar MA, Ahangari Cohan R, Ahmadi K, Asadi Karam MR, Bouzari S. In silico analysis and in vivo assessment of a novel epitope-based vaccine candidate against uropathogenic Escherichia coli. Sci Rep 2020; 10:16258. [PMID: 33004862 PMCID: PMC7530722 DOI: 10.1038/s41598-020-73179-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 09/10/2020] [Indexed: 12/20/2022] Open
Abstract
Uropathogenic Escherichia coli (UPEC) are common pathogens in urinary tract infections (UTIs), which show resistance to antibiotics. Therefore, there is a need for a vaccine to reduce susceptibility to the infection. In the present study, bioinformatics approaches were employed to predict the best B and T-cell epitopes of UPEC virulence proteins to develop a multiepitope vaccine candidate against UPEC. Then, the efficacy of the candidate was studied with and without Freund adjuvant. Using bioinformatics methods, 3 epitope-rich domains of IutA and FimH antigens were selected to construct the fusion. Molecular docking and Molecular dynamics (MD) simulation were employed to investigate in silico interaction between designed vaccine and Toll-like receptor 4 (TLR4). Our results showed that the levels of IgG and IgA antibodies were improved in the serum and mucosal samples of the vaccinated mice, and the IgG responses were maintained for at least 6 months. The fusion protein was also able to enhance the level of cytokines IFN.γ (Th1), IL.4 (Th2), and IL.17. In challenge experiments, all vaccine combinations showed high potency in the protection of the urinary tract even after 6 months post first injection. The present study indicates that the designed candidate is able to evoke strong protective responses which warrant further studies.
Collapse
Affiliation(s)
- Sara Hasanzadeh
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Mehri Habibi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Khadijeh Ahmadi
- Infectious and Tropical Diseases Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
11
|
Kim M, Lee JE, Cho H, Jung HG, Lee W, Seo HY, Lee SH, Ahn DG, Kim SJ, Yu JW, Oh JW. Antiviral efficacy of orally delivered neoagarohexaose, a nonconventional TLR4 agonist, against norovirus infection in mice. Biomaterials 2020; 263:120391. [PMID: 32977259 DOI: 10.1016/j.biomaterials.2020.120391] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/07/2020] [Accepted: 09/16/2020] [Indexed: 12/18/2022]
Abstract
The neoagarohexaose (NA6) is an oligosaccharide that is derived from agarose, the major component of red algae cell walls, by enzymatic hydrolysis. Here we show that NA6 is a noncanonical Toll-like receptor 4 (TLR4) agonist with antiviral activity against norovirus. Its TLR4 activation was dependent on myeloid differentiation factor 2 (MD2) and cluster of differentiation 14 (CD14), leading to interferon-β (IFN-β) and tumor necrosis factor-α (TNF-α) production. This effect was abolished by TLR4 knockdown or knockout in murine macrophages. NA6 inhibited murine norovirus (MNV) replication with an EC50 of 1.5 μM in RAW264.7 cells. It also lowered viral RNA titer in a human hepatocellular carcinoma Huh7-derived cell line harboring a human norovirus subgenomic replicon. The antiviral activity of NA6 was mainly attributed to IFN-β produced through the TLR4-TRIF signaling pathway. NA6-induced TNF-α, which had little effect on norovirus replication per se, primed macrophages to mount greater antiviral innate immune responses when IFN signaling was activated. NA6 boosted the induction of IFN-β in MNV-infected RAW264.7 cells and upregulated IFN-regulatory factor-1, an IFN-stimulated gene. NA6 induced IFN-β expression in the distal ileum with Peyer's patches and oral administration of NA6 reduced MNV loads through activation of TLR4 signaling, highlighting its potential contribution to protective antiviral innate immunity against norovirus.
Collapse
Affiliation(s)
- Minwoo Kim
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Ji-Eun Lee
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Hee Cho
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Hae-Gwang Jung
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Wooseong Lee
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Han Young Seo
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Soung-Hoon Lee
- CK Biotechnology Inc, Engineering Research Park, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Dae-Gyun Ahn
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, 34114, South Korea
| | - Seong-Jun Kim
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, 34114, South Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Program for Leading Universities and Students (PLUS) Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jong-Won Oh
- Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea.
| |
Collapse
|
12
|
Federico S, Pozzetti L, Papa A, Carullo G, Gemma S, Butini S, Campiani G, Relitti N. Modulation of the Innate Immune Response by Targeting Toll-like Receptors: A Perspective on Their Agonists and Antagonists. J Med Chem 2020; 63:13466-13513. [PMID: 32845153 DOI: 10.1021/acs.jmedchem.0c01049] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are a class of proteins that recognize pathogen-associated molecular patterns (PAMPs) and damaged-associated molecular patterns (DAMPs), and they are involved in the regulation of innate immune system. These transmembrane receptors, localized at the cellular or endosomal membrane, trigger inflammatory processes through either myeloid differentiation primary response 88 (MyD88) or TIR-domain-containing adapter-inducing interferon-β (TRIF) signaling pathways. In the last decades, extensive research has been performed on TLR modulators and their therapeutic implication under several pathological conditions, spanning from infections to cancer, from metabolic disorders to neurodegeneration and autoimmune diseases. This Perspective will highlight the recent discoveries in this field, emphasizing the role of TLRs in different diseases and the therapeutic effect of their natural and synthetic modulators, and it will discuss insights for the future exploitation of TLR modulators in human health.
Collapse
Affiliation(s)
- Stefano Federico
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Luca Pozzetti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Alessandro Papa
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| | - Nicola Relitti
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022, University of Siena, via Aldo Moro 2, 53100, Siena, Italy
| |
Collapse
|
13
|
Lee TY, Kim CU, Lee P, Seo SH, Bae EH, Kim YS, Kim SH, Kim DJ. Outer membrane vesicle increases the efficacy of an influenza vaccine in a diet-induced obese mouse model. Immunol Lett 2019; 219:27-33. [PMID: 31901375 DOI: 10.1016/j.imlet.2019.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/20/2019] [Accepted: 12/30/2019] [Indexed: 11/15/2022]
Abstract
Obesity has been associated with increased symptoms and mortality in influenza patients and impaired immune responses to the influenza vaccine. To date, however, there is no effective adjuvant to improve vaccine efficacy for the obese population. To address this issue, we generated a modified outer membrane vesicle with attenuated endotoxicity (fmOMV) and tested its adjuvant effect on the influenza vaccine in comparison with a squalene-based oil-in-water adjuvant (AddaVax) using a diet-induced obese (DIO) mouse model. Although coadministration of fmOMV did not affect neutralizing antibody (Ab) response, it preferentially induced IgG2c antibody response and significantly increased the vaccine-induced T cell response. More importantly, fmOMV conferred significant protection against homologous and heterologous influenza virus challenge, whereas AddaVax showed marginal protection irrespective of the strongest Ab and T cell responses in DIO mice. These results indicate that fmOMV improves the antigen-specific T cell response and the efficacy of an influenza vaccine, suggesting a potential influenza vaccine adjuvant for the obese population.
Collapse
Affiliation(s)
- Tae-Young Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; Department of Biochemistry, Chungnam National University, Daejeon, South Korea
| | - Pureum Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | - Sang-Hwan Seo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Eun-Hye Bae
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Young Sang Kim
- Department of Biochemistry, Chungnam National University, Daejeon, South Korea
| | - Sang-Hyun Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea.
| | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; Department of Biochemistry, Chungnam National University, Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea.
| |
Collapse
|
14
|
Xu ZN, Zheng GD, Wu CB, Jiang XY, Zou SM. Identification of proteins differentially expressed in the gills of grass carp (Ctenopharyngodon idella) after hypoxic stress by two-dimensional gel electrophoresis analysis. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:743-752. [PMID: 30758701 DOI: 10.1007/s10695-018-0599-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/29/2018] [Indexed: 06/09/2023]
Abstract
Two-dimensional gel electrophoresis (2-DE) was combined with liquid chromatography-mass spectrometry (LC-MS/MS) to identify the differential proteomics of grass carp gills after hypoxic stress to better understand the roles of proteins in the hypoxic response and to explore the possible molecular mechanisms. Protein spots were obtained from a hypoxia-stressed group (372 ± 11 individuals) and a control group (406 ± 14 individuals) using the lmage Master 2D Platinum 7.0 analysis software. Fifteen protein spots were expressed differentially in the hypoxia-stressed group and varied significantly after exposure to the hypoxic conditions. In addition, these differential proteins were identified by mass spectrometry and then searched in a database. We found the expression and upregulation of the toll-like receptor 4, ephx1 protein, isocitrate dehydrogenase, L-lactate dehydrogenase, GTP-binding nuclear protein Ran, and glyceraldehyde-3-phosphate dehydrogenase; however, the expression of the keratin type II cytoskeletal 8, type I cytokeratin, ARP3 actin-related protein 3 homolog, thyroid hormone receptor alpha-A, ATP synthase subunit beta, citrate synthase, tropomyosin 2, and tropomyosin 3 were downregulated. Six proteins were found in the hypoxia-inducible factor-1 (HIF-1) signaling pathway. We concluded that the grass carp gill is involved in response processes, including energy generation, metabolic processes, cellular structure, antioxidation, immunity, and signal transduction, to hypoxic stress. To our knowledge, this is the first study to conduct a proteomics analysis of expressed proteins in the gills of grass carp, and this study will help increase the understanding of the molecular mechanisms involved in hypoxic stress responses in fish at the protein level.
Collapse
Affiliation(s)
- Zhan-Ning Xu
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China
- Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China
| | - Guo-Dong Zheng
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China
- Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China
| | - Cheng-Bin Wu
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China
- Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China
| | - Xia-Yun Jiang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China.
- Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China.
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
| | - Shu-Ming Zou
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China.
- Genetics and Breeding Center for Blunt Snout Bream, Ministry of Agriculture, Shanghai Ocean University, Huchenghuan Road 999, Shanghai, 201306, China.
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
15
|
Liu ZF, Chen JL, Li WY, Fan MW, Li YH. FimH as a mucosal adjuvant enhances persistent antibody response and protective efficacy of the anti-caries vaccine. Arch Oral Biol 2019; 101:122-129. [PMID: 30927661 DOI: 10.1016/j.archoralbio.2019.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/07/2019] [Accepted: 02/16/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate whether the recombinant FimH-S.T protein could modulate immune response to anti-caries vaccine in vitro and in vivo. DESIGN Recombinant FimH protein derived from Salmonella was constructed and purified. The expression of dendritic cell maturation markers and cytokines release were performed by flow cytometry, Real-time PCR and ELISA. In addition, BALB/c mice were administered with anti-caries PAc vaccine plus FimH-S.T, antibody responses were evaluated by ELISA. Splenocytes of immunized mice were detected for their proliferative ability in response to in vitro retreatment with PAc antigen by flow cytometry. Caries protection against dental caries formation was also investigated. RESULTS The purified FimH-S.T induced phenotypic maturation of DC2.4 by up-regulating the expression of costimulatory molecules and MHC II, provoked the production and secretion of cytokines via TLR4-dependent signaling pathway in vitro. Furthermore, the mice immunized with the mixture of FimH-S.T and PAc significantly enhanced the PAc-specific antibodies in the serum along with saliva and promoted splenocyte proliferation. Our results also confirmed that PAc+FimH-S.T decreased the caries lesions formation which provided high protective efficacy against dental caries. CONCLUSION Our study demonstrates that recombinant FimH-S.T could enhance specific IgA responses and protection of anti-caries vaccine, possessing mucosal adjuvant ability by activating DC2.4 via TLR4 signaling pathway.
Collapse
Affiliation(s)
- Zhong-Fang Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China
| | - Jun-Lan Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China
| | - Wu-You Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China
| | - Ming-Wen Fan
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China.
| | - Yu-Hong Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan 430079,China.
| |
Collapse
|
16
|
Bae EH, Seo SH, Kim CU, Jang MS, Song MS, Lee TY, Jeong YJ, Lee MS, Park JH, Lee P, Kim YS, Kim SH, Kim DJ. Bacterial Outer Membrane Vesicles Provide Broad-Spectrum Protection against Influenza Virus Infection via Recruitment and Activation of Macrophages. J Innate Immun 2019; 11:316-329. [PMID: 30844806 DOI: 10.1159/000494098] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/16/2018] [Indexed: 12/24/2022] Open
Abstract
Influenza A virus (IAV) poses a constant worldwide threat to human health. Although conventional vaccines are available, their protective efficacy is type or strain specific, and their production is time-consuming. For the control of an influenza pandemic in particular, agents that are immediately effective against a wide range of virus variants should be developed. Although pretreatment of various Toll-like receptor (TLR) ligands have already been reported to be effective in the defense against subsequent IAV infection, the efficacy was limited to specific subtypes, and safety concerns were also raised. In this study, we investigated the protective effect of an attenuated bacterial outer membrane vesicle -harboring modified lipid A moiety of lipopolysaccharide (fmOMV) against IAV infection and the underlying mechanisms. Administration of fmOMV conferred significant protection against a lethal dose of pandemic H1N1, PR8, H5N2, and highly pathogenic H5N1 viruses; this broad antiviral activity was dependent on macrophages but independent of neutrophils. fmOMV induced recruitment and activation of macrophages and elicited type I IFNs. Intriguingly, fmOMV showed a more significant protective effect than other TLR ligands tested in previous reports, without exhibiting any adverse effect. These results show the potential of fmOMV as a prophylactic agent for the defense against influenza virus infection.
Collapse
Affiliation(s)
- Eun-Hye Bae
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Sang Hwan Seo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea
| | - Min Seong Jang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Min-Suk Song
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Republic of Korea
| | - Tae-Young Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Yu-Jin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Moo-Seung Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Jong-Hwan Park
- College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Pureum Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Young Sang Kim
- Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea
| | - Sang-Hyun Kim
- College of Veterinary Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea, .,Department of Biochemistry, Chungnam National University, Daejeon, Republic of Korea, .,University of Science and Technology (UST), Daejeon, Republic of Korea,
| |
Collapse
|
17
|
Sánchez-Ramón S, Conejero L, Netea MG, Sancho D, Palomares Ó, Subiza JL. Trained Immunity-Based Vaccines: A New Paradigm for the Development of Broad-Spectrum Anti-infectious Formulations. Front Immunol 2018; 9:2936. [PMID: 30619296 PMCID: PMC6304371 DOI: 10.3389/fimmu.2018.02936] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
Challenge with specific microbial stimuli induces long lasting epigenetic changes in innate immune cells that result in their enhanced response to a second challenge by the same or unrelated microbial insult, a process referred to as trained immunity. This opens a new avenue in vaccinology to develop Trained Immunity-based Vaccines (TIbV), defined as vaccine formulations that induce training in innate immune cells. Unlike conventional vaccines, which are aimed to elicit only specific responses to vaccine-related antigens, TIbV aim to stimulate broader responses. As trained immunity is generally triggered by pattern recognition receptors (PRRs), TIbV should be formulated with microbial structures containing suitable PRR-ligands. The TIbV concept we describe here may be used for the development of vaccines focused to promote host resistance against a wide spectrum of pathogens. Under the umbrella of trained immunity, a broad protection can be achieved by: (i) increasing the nonspecific effector response of innate immune cells (e.g., monocyte/macrophages) to pathogens, (ii) harnessing the activation state of dendritic cells to enhance adaptive T cell responses to both specific and nonrelated (bystander) antigens. This capacity of TIbV to promote responses beyond their nominal antigens may be particularly useful when conventional vaccines are not available or when multiple coinfections and/or recurrent infections arise in susceptible individuals. As the set of PRR-ligands chosen is essential not only for stimulating trained immunity but also to drive adaptive immunity, the precise design of TIbV will improve with the knowledge on the functional relationship among the different PRRs. While the TIbV concept is emerging, a number of the current anti-infectious vaccines, immunostimulants, and even vaccine adjuvants may already fall in the TIbV category. This may apply to increase immunogenicity of novel vaccine design approaches based on small molecules, like those achieved by reverse vaccinology.
Collapse
Affiliation(s)
- Silvia Sánchez-Ramón
- Department of Clinical Immunology and IdISSC, Hospital Clínico San Carlos, Madrid, Spain.,Department of Immunology, ENT and Ophthalmology, Complutense University School of Medicine, Madrid, Spain
| | | | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department for Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Óscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | | |
Collapse
|
18
|
Wei W, Wan H, Peng X, Zhou H, Lu Y, He Y. Antiviral effects of Ma Huang Tang against H1N1 influenza virus infection in vitro and in an ICR pneumonia mouse model. Biomed Pharmacother 2018; 102:1161-1175. [PMID: 29710534 DOI: 10.1016/j.biopha.2018.03.161] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/25/2018] [Accepted: 03/26/2018] [Indexed: 02/08/2023] Open
Abstract
Ma Huang Tang (MHT), a classical Chinese herbal decoction which has been used in clinic for thousands of years, was very effective in treating the upper respiratory tract infection. But its activity against influenza virus A, the anti-inflammatory effect and the underlying mechanisms have been poorly investigated in previous researches. In this study, the antiviral efficacy of MHT directly inhibiting influenza virus A was investigated in vitro in MDCK cells. In an ICR pneumonia mouse model infected with influenza virus A PR/8/34, MHT (8, 4 and 2 g/kg) were oral administrated for 7 days after viral challenge, to evaluate the effect of MHT on ameliorating viral pneumonia and decipher the underlying mechanisms. The in vitro results showed that MHT possessed antiviral activity with low toxicity. The in vivo assays showed that MHT (8 and 4 g/kg) significantly attenuated lung histopathological changes, decreased lung index, interleukin (IL)-4,5, tumor necrosis factor alpha (TNF-α), CD3+, CD8+ T cell levels, increased IL-2, gamma interferon (IFN-γ), CD4+ T cell levels and CD4+/CD8+ ratio, inhibited toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88) and tumor necrosis factor receptor associated factor 6 (TRAF6) protein levels. All these results demonstrate that MHT can strikingly ameliorate influenza virus A pneumonia in mice, which is associated with the regulating effect of MHT in the imbalance of body's immune function and the MyD88-dependent signaling pathway of TLR4.
Collapse
Affiliation(s)
- Wenyang Wei
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xueqian Peng
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yiyu Lu
- Zhejiang Center for Disease Control and Prevention, Hangzhou 310053, China
| | - Yu He
- Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
19
|
Abdul-Cader MS, Ahmed-Hassan H, Amarasinghe A, Nagy E, Sharif S, Abdul-Careem MF. Toll-like receptor (TLR)21 signalling-mediated antiviral response against avian influenza virus infection correlates with macrophage recruitment and nitric oxide production. J Gen Virol 2017; 98:1209-1223. [PMID: 28613150 DOI: 10.1099/jgv.0.000787] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Cytosine-guanosinedeoxynucleotide (CpG) DNA can be used for the stimulation of the toll-like receptor (TLR)21 signalling pathway in avian species which ultimately leads to up-regulation of gene transcription for pro-inflammatory molecules including nitric oxide and recruitment of innate immune cells. The objective of this study was to determine the antiviral effect of NO, produced in response to in ovo delivery of CpG DNA, against avian influenza virus (AIV) infection. We found that when CpG DNA is delivered at embryo day (ED)18 in ovo and subsequently challenged with H4N6 AIV at ED19 pre-hatch and day 1 post-hatching, CpG DNA reduces H4N6 AIV replication associated with enhanced NO production and macrophage recruitment in lungs. In vitro, we showed that NO originating from macrophages is capable of eliciting an antiviral response against H4N6 AIV infection. This study provides insights into the mechanisms of CpG DNA-mediated antiviral response, particularly against AIV infection in avian species.
Collapse
Affiliation(s)
- Mohamed Sarjoon Abdul-Cader
- Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Centre 2C53, 3330 Hospital Drive NW, Calgary, AB, Canada, AB T2N 4N1
| | - Hanaa Ahmed-Hassan
- Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Centre 2C53, 3330 Hospital Drive NW, Calgary, AB, Canada, AB T2N 4N1
| | - Aruna Amarasinghe
- Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Centre 2C53, 3330 Hospital Drive NW, Calgary, AB, Canada, AB T2N 4N1
| | - Eva Nagy
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada, ON N1G 2W1
| | - Shayan Sharif
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada, ON N1G 2W1
| | - Mohamed Faizal Abdul-Careem
- Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Centre 2C53, 3330 Hospital Drive NW, Calgary, AB, Canada, AB T2N 4N1
| |
Collapse
|
20
|
Callegan MC, Parkunan SM, Randall CB, Coburn PS, Miller FC, LaGrow AL, Astley RA, Land C, Oh SY, Schneewind O. The role of pili in Bacillus cereus intraocular infection. Exp Eye Res 2017; 159:69-76. [PMID: 28336259 PMCID: PMC5492386 DOI: 10.1016/j.exer.2017.03.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/09/2017] [Accepted: 03/16/2017] [Indexed: 12/15/2022]
Abstract
Bacterial endophthalmitis is a potentially blinding intraocular infection. The bacterium Bacillus cereus causes a devastating form of this disease which progresses rapidly, resulting in significant inflammation and loss of vision within a few days. The outer surface of B. cereus incites the intraocular inflammatory response, likely through interactions with innate immune receptors such as TLRs. This study analyzed the role of B. cereus pili, adhesion appendages located on the bacterial surface, in experimental endophthalmitis. To test the hypothesis that the presence of pili contributed to intraocular inflammation and virulence, we analyzed the progress of experimental endophthalmitis in mouse eyes infected with wild type B. cereus (ATCC 14579) or its isogenic pilus-deficient mutant (ΔbcpA-srtD-bcpB or ΔPil). One hundred CFU were injected into the mid-vitreous of one eye of each mouse. Infections were analyzed by quantifying intraocular bacilli and retinal function loss, and by histology from 0 to 12 h postinfection. In vitro growth and hemolytic phenotypes of the infecting strains were also compared. There was no difference in hemolytic activity (1:8 titer), motility, or in vitro growth (p > 0.05, every 2 h, 0-18 h) between wild type B. cereus and the ΔPil mutant. However, infected eyes contained greater numbers of wild type B. cereus than ΔPil during the infection course (p ≤ 0.05, 3-12 h). Eyes infected with wild type B. cereus experienced greater losses in retinal function than eyes infected with the ΔPil mutant, but the differences were not always significant. Eyes infected with ΔPil or wild type B. cereus achieved similar degrees of severe inflammation. The results indicated that the intraocular growth of pilus-deficient B. cereus may have been better controlled, leading to a trend of greater retinal function in eyes infected with the pilus-deficient strain. Although this difference was not enough to significantly alter the severity of the inflammatory response, these results suggest a potential role for pili in protecting B. cereus from clearance during the early stages of endophthalmitis, which is a newly described virulence mechanism for this organism and this infection.
Collapse
Affiliation(s)
- Michelle C. Callegan
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA,Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, 950 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA,Dean A. McGee Eye Institute, Oklahoma City Oklahoma USA,Corresponding author: DMEI PA-418, 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA. Phone: (405) 271-3674, Fax: (405) 271-8128,
| | - Salai Madhumathi Parkunan
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, 950 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA
| | - C. Blake Randall
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA
| | - Phillip S. Coburn
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA
| | - Frederick C. Miller
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 950 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA
| | - Austin L. LaGrow
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA
| | - Roger A. Astley
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA
| | - Craig Land
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA
| | - So-Young Oh
- Department of Microbiology, University of Chicago, 920 East 58
| | - Olaf Schneewind
- Department of Microbiology, University of Chicago, 920 East 58
| |
Collapse
|
21
|
Incorporation of a bi-functional protein FimH enhances the immunoprotection of chitosan-pVP1 vaccine against coxsackievirus B3-induced myocarditis. Antiviral Res 2017; 140:121-132. [DOI: 10.1016/j.antiviral.2017.01.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 01/12/2023]
|
22
|
Lee TY, Kim CU, Bae EH, Seo SH, Jeong DG, Yoon SW, Chang KT, Kim YS, Kim SH, Kim DJ. Outer membrane vesicles harboring modified lipid A moiety augment the efficacy of an influenza vaccine exhibiting reduced endotoxicity in a mouse model. Vaccine 2016; 35:586-595. [PMID: 28024958 PMCID: PMC7115551 DOI: 10.1016/j.vaccine.2016.12.025] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/24/2016] [Accepted: 12/09/2016] [Indexed: 12/20/2022]
Abstract
Influenza is an acute respiratory disease and a major health problem worldwide. Since mucosal immunity plays a critical role in protection against influenza virus infection, mucosal immunization is considered a promising vaccination route. However, except for live-attenuated vaccines, there are no effective killed or recombinant mucosal influenza vaccines to date. Outer membrane vesicles (OMVs) are nano-sized vesicles produced by gram-negative bacteria, and contain various bacterial components capable of stimulating the immune system of the host. We generated an OMV with low endotoxicity (fmOMV) by modifying the structure of the lipid A moiety of lipopolysaccharide and investigated its effect as an intranasal vaccine adjuvant in an influenza vaccine model. In this model, fmOMV exhibited reduced toll-like receptor 4-stimulating activity and attenuated endotoxicity compared to that of native OMV. Intranasal injection of the vaccine antigen with fmOMV significantly increased systemic antibody and T cell responses, mucosal IgA levels, and the frequency of lung-resident influenza-specific T cells. In addition, the number of antigen-bearing CD103+ dendritic cells in the mediastinal lymph nodes was significantly increased after fmOMV co-administration. Notably, the mice co-immunized with fmOMV showed a significantly higher protection rate against challenge with a lethal dose of homologous or heterologous influenza viruses without adverse effects. These results show the potential of fmOMV as an effective mucosal adjuvant for intranasal vaccines.
Collapse
Affiliation(s)
- Tae-Young Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Chang-Ung Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; Department of Biochemistry, Chungnam National University, Daejeon, South Korea
| | - Eun-Hye Bae
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Sang-Hwan Seo
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Dae Gwin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | - Sun-Woo Yoon
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea
| | - Kyu-Tae Chang
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea
| | - Young Sang Kim
- Department of Biochemistry, Chungnam National University, Daejeon, South Korea
| | - Sang-Hyun Kim
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea.
| | - Doo-Jin Kim
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, South Korea; Department of Biochemistry, Chungnam National University, Daejeon, South Korea; University of Science and Technology (UST), Daejeon, South Korea.
| |
Collapse
|
23
|
Shah M, Anwar MA, Kim JH, Choi S. Advances in Antiviral Therapies Targeting Toll-like Receptors. Expert Opin Investig Drugs 2016; 25:437-53. [DOI: 10.1517/13543784.2016.1154040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
24
|
Habibi M, Asadi Karam MR, Bouzari S. Transurethral instillation with fusion protein MrpH.FimH induces protective innate immune responses against uropathogenic Escherichia coli and Proteus mirabilis. APMIS 2016; 124:444-52. [PMID: 26918627 DOI: 10.1111/apm.12523] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/12/2016] [Indexed: 12/25/2022]
Abstract
Urinary tract infections (UTIs) are among the most common infections in human. Innate immunity recognizes pathogen-associated molecular patterns (PAMPs) by Toll-like receptors (TLRs) to activate responses against pathogens. Recently, we demonstrated that MrpH.FimH fusion protein consisting of MrpH from Proteus mirabilis and FimH from Uropathogenic Escherichia coli (UPEC) results in the higher immunogenicity and protection, as compared with FimH and MrpH alone. In this study, we evaluated the innate immunity and adjuvant properties induced by fusion MrpH.FimH through in vitro and in vivo methods. FimH and MrpH.FimH were able to induce significantly higher IL-8 and IL-6 responses than untreated or MrpH alone in cell lines tested. The neutrophil count was significantly higher in the fusion group than other groups. After 6 h, IL-8 and IL-6 production reached a peak, with a significant decline at 24 h post-instillation in both bladder and kidney tissues. Mice instilled with the fusion and challenged with UPEC or P. mirabilis showed a significant decrease in the number of bacteria in bladder and kidney compared to control mice. The results of these studies demonstrate that the use of recombinant fusion protein encoding TLR-4 ligand represents an effective vaccination strategy that does not require the use of a commercial adjuvant. Furthermore, MrpH.FimH was presented as a promising vaccine candidate against UTIs caused by UPEC and P. mirabilis.
Collapse
Affiliation(s)
- Mehri Habibi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Saeid Bouzari
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
25
|
Dual ligand/receptor interactions activate urothelial defenses against uropathogenic E. coli. Sci Rep 2015; 5:16234. [PMID: 26549759 PMCID: PMC4637824 DOI: 10.1038/srep16234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/12/2015] [Indexed: 12/26/2022] Open
Abstract
During urinary tract infection (UTI), the second most common bacterial infection, dynamic interactions take place between uropathogenic E. coli (UPEC) and host urothelial cells. While significant strides have been made in the identification of the virulence factors of UPEC, our understanding of how the urothelial cells mobilize innate defenses against the invading UPEC remains rudimentary. Here we show that mouse urothelium responds to the adhesion of type 1-fimbriated UPEC by rapidly activating the canonical NF-κB selectively in terminally differentiated, superficial (umbrella) cells. This activation depends on a dual ligand/receptor system, one between FimH adhesin and uroplakin Ia and another between lipopolysaccharide and Toll-like receptor 4. When activated, all the nuclei (up to 11) of a multinucleated umbrella cell are affected, leading to significant amplification of proinflammatory signals. Intermediate and basal cells of the urothelium undergo NF-κB activation only if the umbrella cells are detached or if the UPEC persistently express type 1-fimbriae. Inhibition of NF-κB prevents the urothelium from clearing the intracellular bacterial communities, leading to prolonged bladder colonization by UPEC. Based on these data, we propose a model of dual ligand/receptor system in innate urothelial defenses against UPEC.
Collapse
|
26
|
Peng XQ, Zhou HF, Zhang YY, Yang JH, Wan HT, He Y. Antiviral effects of Yinhuapinggan granule against influenza virus infection in the ICR mice model. J Nat Med 2015; 70:75-88. [PMID: 26439479 DOI: 10.1007/s11418-015-0939-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/09/2015] [Indexed: 01/12/2023]
Abstract
Yinhuapinggan granule (YHPG), a Chinese medicine granule based on Ma-Huang-Tang (Ephedra Decoction) and the clinical experience of Professor Wan Haitong, is used in traditional Chinese medicine (TCM) for the treatment of colds, influenza, fever, inflammation and cough. This study investigated the antiviral effects of YHPG on the production of inflammatory cytokines in influenza virus (IFV)-infected mice and evaluated the effect of YHPG on the expression of NF-κB p65 and the level of key signaling molecules in the TLR4 signaling pathway. ICR mice were orally administrated YHPG at doses of 7.5, 15 and 30 g kg(-1) day(-1) for 2 or 6 days after IFV infection. On days 3 and 7 after infection, YHPG (15 g/kg and 30 g/kg) significantly increased levels of interleukin (IL)-2 and interferon gamma and decreased levels of IL-4, IL-5 and tumor necrosis factor (TNF) in serum compared with the IFV control group. Furthermore, the expression of TLR4, MyD88, TRAF6 and NF-κB p65 at the mRNA and protein level was significantly lower in the YHPG (15 and 30 g/kg) treatment groups than in the IFV control group. These results suggest that YHPG has antiviral effects in IFV-infected mice, which is associated with the inhibition of the TLR4-MyD88-TRAF6 signaling pathway and the expression of NF-κB p65.
Collapse
Affiliation(s)
- Xue-qian Peng
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hui-fen Zhou
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yu-yan Zhang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jie-hong Yang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Hai-tong Wan
- Institute of Cardio-Cerebrovascular Diseases, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, China.
| | - Yu He
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, China.
| |
Collapse
|
27
|
Choi HG, Kim WS, Back YW, Kim H, Kwon KW, Kim JS, Shin SJ, Kim HJ. Mycobacterium tuberculosis RpfE promotes simultaneous Th1- and Th17-type T-cell immunity via TLR4-dependent maturation of dendritic cells. Eur J Immunol 2015; 45:1957-71. [PMID: 25907170 DOI: 10.1002/eji.201445329] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/31/2015] [Accepted: 04/21/2015] [Indexed: 11/12/2022]
Abstract
Reciprocal induction of the Th1 and Th17 immune responses is essential for optimal protection against Mycobacterium tuberculosis (Mtb); however, only a few Mtb antigens are known to fulfill this task. A functional role for resuscitation-promoting factor (Rpf) E, a latency-associated member of the Rpf family, in promoting naïve CD4(+) T-cell differentiation toward both Th1 and Th17 cell fates through interaction with dendritic cells (DCs) was identified in this study. RpfE induces DC maturation by increasing expression of surface molecules and the production of IL-6, IL-1β, IL-23p19, IL-12p70, and TNF-α but not IL-10. This induction is mediated through TLR4 binding and subsequent activation of ERK, p38 MAPKs, and NF-κB signaling. RpfE-treated DCs effectively caused naïve CD4(+) T cells to secrete IFN-γ, IL-2, and IL-17A, which resulted in reciprocal expansions of the Th1 and Th17 cell response along with activation of T-bet and RORγt but not GATA-3. Furthermore, lung and spleen cells from Mtb-infected WT mice but not from TLR4(-/-) mice exhibited Th1 and Th17 polarization upon RpfE stimulation. Taken together, our data suggest that RpfE has the potential to be an effective Mtb vaccine because of its ability to activate DCs that simultaneously induce both Th1- and Th17-polarized T-cell expansion.
Collapse
Affiliation(s)
- Han-Gyu Choi
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea.,Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Woo Sik Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Yong Woo Back
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong-Seok Kim
- Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hwa-Jung Kim
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea.,Infection Signaling Network Research Center, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
28
|
Thapa S, Nagy E, Abdul-Careem MF. In ovo delivery of Toll-like receptor 2 ligand, lipoteichoic acid induces pro-inflammatory mediators reducing post-hatch infectious laryngotracheitis virus infection. Vet Immunol Immunopathol 2015; 164:170-8. [PMID: 25764942 DOI: 10.1016/j.vetimm.2015.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 02/12/2015] [Accepted: 02/13/2015] [Indexed: 01/07/2023]
Abstract
Toll-like receptor (TLR) ligands are pathogen associated molecular patterns (PAMPs) recognized by the TLRs resulting in induction of host innate immune responses. One of the PAMPs that binds to TLR2 and cluster of differentiation (CD) 14 is lipotechoic acid (LTA), which activates downstream signals culminating in the release of pro-inflammatory cytokines. In this study, we investigated whether in ovo LTA delivery leads to the induction of antiviral responses against post-hatch infectious laryngotracheitis virus (ILTV) infection. We first delivered the LTA into embryo day (ED)18 eggs via in ovo route so that the compound is available at the respiratory mucosa. Then the LTA treated and control ED18 eggs were allowed to hatch and the hatched chicken was infected with ILTV intratracheally on the day of hatch. We found that in ovo delivered LTA reduces ILTV infection post-hatch. We also found that in ovo delivery of LTA significantly increases mRNA expression of pro-inflammatory mediators in pre-hatch embryo lungs as well as mononuclear cell infiltration, predominantly macrophages, in lung of post-hatch chickens. Altogether, the data suggest that in ovo delivered LTA could be used to reduce ILTV infection in newly hatched chickens.
Collapse
Affiliation(s)
- S Thapa
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, Canada AB T2N 2Z6
| | - E Nagy
- Department of Pathobiology, University of Guelph, Guelph, Canada
| | - M F Abdul-Careem
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Health Research Innovation Center 2C53, 3330 Hospital Drive NW, Calgary, Canada AB T2N 2Z6.
| |
Collapse
|
29
|
Intratracheal administration of influenza virus is superior to intranasal administration as a model of acute lung injury. J Virol Methods 2014; 209:116-20. [PMID: 25239366 DOI: 10.1016/j.jviromet.2014.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 08/27/2014] [Accepted: 09/09/2014] [Indexed: 12/31/2022]
Abstract
Infection of mice with human or murine adapted influenza A viruses results in a severe pneumonia. However, the results of studies from different laboratories show surprising variability, even in genetically similar strains. Differences in inoculum size related to the route of viral delivery (intranasal vs. intratracheal) might explain some of this variability. To test this hypothesis, mice were infected intranasally or intratracheally with different doses of influenza A virus (A/WSN/33 [H1N1]). Daily weights, a requirement for euthanasia, viral load in the lungs and brains, inflammatory cytokines, wet-to-dry ratio, total protein and histopathology of the infected mice were examined. With all doses of influenza tested, intranasal delivery resulted in less severe lung injury, as well as smaller and more variable viral loads in the lungs when compared with intratracheal delivery. Virus was not detected in the brain following either method of delivery. It is concluded that compared to intranasal infection, intratracheal infection with influenza A virus is a more reliable method to deliver virus to the lungs.
Collapse
|
30
|
Barjesteh N, Behboudi S, Brisbin JT, Villanueva AI, Nagy É, Sharif S. TLR ligands induce antiviral responses in chicken macrophages. PLoS One 2014; 9:e105713. [PMID: 25165812 PMCID: PMC4148336 DOI: 10.1371/journal.pone.0105713] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 07/23/2014] [Indexed: 11/30/2022] Open
Abstract
Chicken macrophages express several receptors for recognition of pathogens, including Toll-like receptors (TLRs). TLRs bind to pathogen-associated molecular patterns (PAMPs) derived from bacterial or viral pathogens leading to the activation of macrophages. Macrophages play a critical role in immunity against viruses, including influenza viruses. The present study was designed to test the hypothesis that treatment of chicken macrophages with TLR ligands reduces avian influenza replication. Furthermore, we sought to study the expression of some of the key mediators involved in the TLR-mediated antiviral responses of macrophages. Chicken macrophages were treated with the TLR2, 3, 4, 7 and 21 ligands, Pam3CSK4, poly(I:C), LPS, R848 and CpG ODN, respectively, at different doses and time points pre- and post-H4N6 avian influenza virus (AIV) infection. The results revealed that pre-treatment of macrophages with Pam3CSK4, LPS and CpG ODN reduced the replication of AIV in chicken macrophages. In addition, the relative expression of genes involved in inflammatory and antiviral responses were quantified at 3, 8 and 18 hours post-treatment with the TLR2, 4 and 21 ligands. Pam3CSK4, LPS and CpG ODN increased the expression of interleukin (IL)-1β, interferon (IFN)-γ, IFN-β and interferon regulatory factor (IFR) 7. The expression of these genes correlated with the reduction of viral replication in macrophages. These results shed light on the process of immunity to AIV in chickens.
Collapse
Affiliation(s)
- Neda Barjesteh
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Shahriar Behboudi
- The Pirbright Institute, Compton Laboratory, Newbury, United Kingdom
| | | | | | - Éva Nagy
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
| | - Shayan Sharif
- Department of Pathobiology, University of Guelph, Guelph, Ontario, Canada
- * E-mail:
| |
Collapse
|
31
|
Morales-Nebreda L, Mutlu GM, Scott Budinger GR, Radigan KA. Loss of TLR4 does not prevent influenza A-induced mortality. Am J Respir Crit Care Med 2014; 189:1280-1. [PMID: 24832747 DOI: 10.1164/rccm.201401-0193le] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
32
|
Zhang M, Luo F, Zhang Y, Wang L, Lin W, Yang M, Hu D, Wu X, Chu Y. Pseudomonas aeruginosa mannose-sensitive hemagglutinin promotes T-cell response via toll-like receptor 4-mediated dendritic cells to slow tumor progression in mice. J Pharmacol Exp Ther 2014; 349:279-87. [PMID: 24623801 DOI: 10.1124/jpet.113.212316] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pseudomonas aeruginosa-mannose-sensitive hemagglutinin (PA-MSHA) as a drug may kill tumor cells and has been used clinically. However, the antitumor immune response of PA-MSHA is not completely understood. In this study, we found that treating Lewis lung carcinoma (3LL)-bearing mice with PA-MSHA plus 3LL antigen led to slower tumor progression and longer survival. After PA-MSHA treatment, T-cell number and dendritic cell maturation were both increased significantly at the tumor site. In addition, PA-MSHA in vitro stimulation resulted in the maturation of bone marrow-derived dendritic cells (BMDCs) from naive mice, showing higher costimulatory molecule expression, more cytokine secretion, lower endocytic activity, and stronger capacity to enhance T-cell activation. Toll-like receptor (TLR)4 but not TLR2 was required in the maturation process. More importantly, PA-MSHA-induced DCs were essential for PA-MSHA to enhance activation, expansion, and interferon (IFN)-γ secretion of TLR4-mediated T cells, which play a role in the antitumor effect of PA-MSHA. Thus, this study reveals PA-MSHA as a novel TLR4 agonist that elicits antitumor immune response to slow tumor progression.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, Department of Immunology, School of Basic Medical Sciences and Biotherapy Research Center (M.Z., F.L., Y.Z., L.W., W.L., Y.C.) and Department of General Surgery, Zhongshan Hospital (M.Y.), Fudan University, Shanghai, China; and Beijing Wanter Biopharmaceutical Co., Ltd., Huairou Yanqi Economic-Technical Development Area, Beijing, China (D.H., X.W.)
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lai R, Jeyanathan M, Shaler CR, Damjanovic D, Khera A, Horvath C, Ashkar AA, Xing Z. Restoration of innate immune activation accelerates Th1-cell priming and protection following pulmonary mycobacterial infection. Eur J Immunol 2014; 44:1375-86. [PMID: 24519467 DOI: 10.1002/eji.201344300] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/12/2014] [Accepted: 02/06/2014] [Indexed: 02/06/2023]
Abstract
The immune mechanisms underlying delayed induction of Th1-type immunity in the lungs following pulmonary mycobacterial infection remain poorly understood. We have herein investigated the underlying immune mechanisms for such delayed responses and whether a selected innate immune-modulating strategy can accelerate Th1-type responses. We have found that, in the early stage of pulmonary infection with attenuated Mycobacterium tuberculosis (M.tb H37Ra), the levels of infection in the lung continue to increase logarithmically until days 14 and 21 postinfection in C57BL/6 mice. The activation of innate immune responses, particularly DCs, in the lung is delayed. This results in a delay in the subsequent downstream immune responses including the migration of antigen-bearing DCs to the draining lymph node (dLN), the Th1-cell priming in dLN, and the recruitment of Th1 cells to the lung. However, single lung mucosal exposure to the TLR agonist FimH postinfection is able to accelerate protective Th1-type immunity via facilitating DC migration to the lung and draining lymph nodes, enhancing DC antigen presentation and Th1-cell priming. These findings hold implications for the development of immunotherapeutic and vaccination strategies and suggest that enhancement of early innate immune activation is a viable option for improving Th1-type immunity against pulmonary mycobacterial diseases.
Collapse
Affiliation(s)
- Rocky Lai
- McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Mifsud EJ, Tan ACL, Jackson DC. TLR Agonists as Modulators of the Innate Immune Response and Their Potential as Agents Against Infectious Disease. Front Immunol 2014; 5:79. [PMID: 24624130 PMCID: PMC3939722 DOI: 10.3389/fimmu.2014.00079] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/13/2014] [Indexed: 12/28/2022] Open
Abstract
Immunotherapies that can either activate or suppress innate immune responses are being investigated as treatments against infectious diseases and the pathology they can cause. The objective of these therapies is to elicit protective immune responses thereby limiting the harm inflicted by the pathogen. The Toll-like receptor (TLR) signaling pathway plays critical roles in numerous host immune defenses and has been identified as an immunotherapeutic target against the consequences of infectious challenge. This review focuses on some of the recent advances being made in the development of TLR-ligands as potential prophylactic and/or therapeutic agents.
Collapse
Affiliation(s)
- Edin J. Mifsud
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Amabel C. L. Tan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - David C. Jackson
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
35
|
The effects of administration of ligands for Toll-like receptor 4 and 21 against Marek's disease in chickens. Vaccine 2014; 32:1932-8. [PMID: 24530927 DOI: 10.1016/j.vaccine.2014.01.082] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 01/05/2014] [Accepted: 01/30/2014] [Indexed: 12/11/2022]
Abstract
Ligands for Toll-like receptors (TLRs) are known to stimulate immune responses, leading to protection against bacterial and viral pathogens. Here, we aimed to examine the effects of various TLR ligands on the development of Marek's disease in chickens. Specific-pathogen free chickens were treated with a series of TLR ligands that interact with TLR3, TLR9 and TLR21. In a pilot study, it was determined that TLR4 and TLR21 ligands are efficacious, in that they could reduce the incidence of Marek's disease tumors in infected birds. Hence, in a subsequent study, chickens were treated with lipopolysaccharide (LPS) as a TLR4 and CpG oligodeoxynucleotides (ODN) as TLR21 agonists before being challenged with the RB1B strain of Marek's disease virus (MDV) via the respiratory route. The results demonstrated that the administration of LPS or CpG ODN, but not PBS or non-CpG ODN, delayed disease onset and reduced MDV genome copy number in the spleens of infected chickens. Taken together, our data demonstrate that TLR4 and 21 agonists modulate anti-virus innate immunity including cytokine responses in MD-infected chicken and this response can only delay, but not inhibit, disease progression.
Collapse
|
36
|
Tsuge M, Oka T, Yamashita N, Saito Y, Fujii Y, Nagaoka Y, Yashiro M, Tsukahara H, Morishima T. Gene expression analysis in children with complex seizures due to influenza A(H1N1)pdm09 or rotavirus gastroenteritis. J Neurovirol 2014; 20:73-84. [PMID: 24464411 DOI: 10.1007/s13365-013-0231-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/11/2013] [Accepted: 12/26/2013] [Indexed: 10/25/2022]
Abstract
Viral infections have been implicated as a cause of complex seizures in children. The pathogenic differences in complex seizures due to influenza A(H1N1)pdm09 or rotavirus gastroenteritis remain unclear. This study analyzed the gene expression profiles in the peripheral whole blood from pediatric patients with complex seizures due to influenza A(H1N1)pdm09 or rotavirus gastroenteritis. The gene expression profiles of ten patients (five with seizures and five without) with influenza A(H1N1)pdm09 and six patients (three with seizures and three without) with rotavirus gastroenteritis were examined. Gene expression profiles in the whole blood were different in complex seizures due to influenza A(H1N1)pdm09 or rotavirus gastroenteritis. Transcripts related to the immune response were significantly differentially expressed in complex seizures with influenza A(H1N1)pdm09, and transcripts related to the stress response were significantly differentially expressed in complex seizures with rotavirus gastroenteritis. Pathway analysis showed that the mitogen-activated protein kinases in the T cell receptor signaling pathway were activated in complex seizures due to influenza A(H1N1)pdm09. Dysregulation of the genes related to immune response or stress response could contribute to the pathogenic differences of the complex seizures due to influenza A(H1N1)pdm09 or rotavirus gastroenteritis.
Collapse
Affiliation(s)
- Mitsuru Tsuge
- Department of Pediatrics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan,
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wu CCN, Crain B, Yao S, Sabet M, Lao FS, Tawatao RI, Chan M, Smee DF, Julander JG, Cottam HB, Guiney DG, Corr M, Carson DA, Hayashi T. Innate immune protection against infectious diseases by pulmonary administration of a phospholipid-conjugated TLR7 ligand. J Innate Immun 2013; 6:315-24. [PMID: 24192551 DOI: 10.1159/000355217] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 08/23/2013] [Indexed: 12/21/2022] Open
Abstract
Pulmonary administration of Toll-like receptor (TLR) ligands protects hosts from inhaled pathogens. However, systemic side effects induced by TLR stimulation limit clinical development. Here, a small-molecule TLR7 ligand conjugated with phospholipid, 1V270 (also designated TMX201), was tested for innate immune activation and its ability to prevent pulmonary infection in mice. We hypothesized that phospholipid conjugation would increase internalization by immune cells and localize the compound in the lungs, thus avoiding side effects due to systemic cytokine release. Pulmonary 1V270 administration increased innate cytokines and chemokines in bronchial alveolar lavage fluids, but neither caused systemic induction of cytokines nor B cell proliferation in distant lymphoid organs. 1V270 activated pulmonary CD11c+ dendritic cells, which migrated to local lymph nodes. However, there was minimal cell infiltration into the pulmonary parenchyma. Prophylactic administration of 1V270 significantly protected mice from lethal infection with Bacillus anthracis, Venezuelan equine encephalitis virus and H1N1 influenza virus. The maximum tolerated dose of 1V270 by pulmonary administration was 75 times the effective therapeutic dose. Therefore, pulmonary 1V270 treatment can protect the host from different infectious agents by stimulating local innate immune responses while exhibiting an excellent safety profile.
Collapse
Affiliation(s)
- Christina C N Wu
- Rebecca and John Moores UCSD Cancer Center, University of California San Diego, La Jolla, Calif., USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lee W, Lee SH, Ahn DG, Cho H, Sung MH, Han SH, Oh JW. The antiviral activity of poly-γ-glutamic acid, a polypeptide secreted by Bacillus sp., through induction of CD14-dependent type I interferon responses. Biomaterials 2013; 34:9700-8. [PMID: 24016850 PMCID: PMC7112489 DOI: 10.1016/j.biomaterials.2013.08.067] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 08/21/2013] [Indexed: 02/06/2023]
Abstract
Poly-γ-glutamic acid (γ-PGA) is an anionic polypeptide secreted by Bacillus sp. that has been shown to activate immune cells through interactions with toll-like receptor 4 (TLR4). However, its ability to induce the type I interferon (IFN) response has not yet been characterized. Here, we demonstrate that γ-PGA induces type I IFN signaling pathway via the TLR4 signaling pathway. The induction required both myeloid differentiation factor 2 (MD2) and the pattern-recognition receptor CD14, which are two TLR4-associated accessory proteins. The γ-PGA with high molecular weights (2000 and 5000 kDa) was able to activate the subsequent signals through TLR4/MD2 to result in dimerization of IRF-3, a transcription factor required for IFN gene expression, leading to increases in mRNA levels of the type I IFN-response genes, 2′–5′ OAS and ISG56. Moreover, γ-PGA (2000 kDa) displayed an antiviral activity against SARS coronavirus and hepatitis C virus. Our results identify high-molecular weight γ-PGA as a TLR4 ligand and demonstrate that γ-PGA requires both CD14 and MD2 for the activation of type I IFN responses. Our results suggest that the microbial biopolymer γ-PGA may have therapeutic potential against a broad range of viruses sensitive to type I IFNs.
Collapse
Affiliation(s)
- Wooseong Lee
- Department of Biotechnology and Center for Protein Function Control, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
39
|
Mathieu C, Rioux G, Dumas MC, Leclerc D. Induction of innate immunity in lungs with virus-like nanoparticles leads to protection against influenza and Streptococcus pneumoniae challenge. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:839-48. [PMID: 23499666 DOI: 10.1016/j.nano.2013.02.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 01/24/2013] [Accepted: 02/19/2013] [Indexed: 11/20/2022]
Abstract
UNLABELLED Nanoparticles composed of the coat protein of a plant virus (papaya mosaic virus; PapMV) and a single-stranded RNA (ssRNA) trigger a strong innate immune stimulation in the lungs of the animals a few hours following instillation. A rapid recruitment of neutrophils, monocytes/macrophages and lymphocytes follows. This treatment was able to provide protection to an influenza challenge that lasts at least 5 days. Protection could be recalled for longer periods by repeating the instillations once per week for more than 10 weeks. The treatment also conferred protection to a lethal challenge with Streptococcus pneumoniae--the major cause of bacterial pneumonia. Finally, we also showed that the nanoparticles could be used to treat mice infected with influenza and significantly decrease morbidity. These data strengthen the potential for using PapMV nanoparticles as non-specific inducers of the innate immune response in lungs during viral pandemics or to combat bioterrorist attack. FROM THE CLINICAL EDITOR In this study, virus-like nanoparticles were utilized to induce innate immune responses in a mouse model. They were also demonstrated to provide enhanced immune responses during actual pneumonia and ongoing viral infection. Strategies like this may become very helpful in human applications, including bioterrorism countermeasures.
Collapse
Affiliation(s)
- Claudia Mathieu
- Department of Microbiology, Infectiology and Immunology, Infectious Disease Research Centre/CHU de Québec, Laval University, Laurier, Quebec City, PQ, Canada
| | | | | | | |
Collapse
|
40
|
Current world literature. Curr Opin Infect Dis 2012; 25:718-28. [PMID: 23147811 DOI: 10.1097/qco.0b013e32835af239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Tan ACL, Mifsud EJ, Zeng W, Edenborough K, McVernon J, Brown LE, Jackson DC. Intranasal administration of the TLR2 agonist Pam2Cys provides rapid protection against influenza in mice. Mol Pharm 2012; 9:2710-8. [PMID: 22823162 DOI: 10.1021/mp300257x] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The protective role played by the innate immune system during early stages of infection suggests that compounds which stimulate innate responses could be used as antimicrobial or antiviral agents. In this study, we demonstrate that the Toll-like receptor-2 agonist Pam2Cys, when administered intranasally, triggers a cascade of inflammatory and innate immune signals, acting as an immunostimulant by attracting neutrophils and macrophages and inducing secretion of IL-2, IL-6, IL-10, IFN-γ, MCP-1 and TNF-α. These changes provide increased resistance against influenza A virus challenge and also reduce the potential for transmission of infection. Pam2Cys treatment also reduced weight loss and lethality associated with virulent influenza virus infection in a Toll-like receptor-2-dependent manner. Treatment did not affect the animals' ability to generate an adaptive immune response, measured by the induction of functional influenza A virus-specific CD8(+) T cells following exposure to virus. Because this compound demonstrates efficacy against distinct strains of influenza, it could be a candidate for development as an agent against influenza and possibly other respiratory pathogens.
Collapse
Affiliation(s)
- Amabel C L Tan
- Department of Microbiology & Immunology, The University of Melbourne, Parkville 3010, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
42
|
Nara JM, Pimenta DC, Abe CM, Abreu PAE, Moraes CTP, Freitas NC, Elias WP, Piazza RMF. Low-molecular mass comparative proteome of four atypical enteropathogenic Escherichia coli isolates showing different adherence patterns. Comp Immunol Microbiol Infect Dis 2012; 35:539-49. [PMID: 22768807 DOI: 10.1016/j.cimid.2012.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 06/04/2012] [Accepted: 06/10/2012] [Indexed: 11/19/2022]
Abstract
Atypical enteropathogenic Escherichia coli (aEPEC) are heterogeneous in terms of serotypes, adherence patterns and the presence of non-locus of enterocyte effacement virulence factors. In this study, the low-molecular mass proteomes of four representative aEPEC, comprising three different adhesion phenotypes (localized-like, aggregative and diffuse) and one non-adherent isolate, were analyzed and compared by 2D gel electrophoresis and LC-MS/MS. By mass spectrometry, a total of 59 proteins were identified according to their annotated function, with most of them being involved in metabolism, protection, and transport; some of them still classified as hypothetical proteins. Thus, in this comparative proteomic analysis of low-molecular mass extracted proteins from different aEPEC isolates, the proteins identified are mainly involved in key metabolic pathways. Also, the majority of the hypothetical and filamentous proteins identified in the isolates studied are products of genes originally identified in the genome of enterohemorrhagic E. coli.
Collapse
Affiliation(s)
- Júlia M Nara
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, 05503-900, São Paulo, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Damjanovic D, Small CL, Jeyananthan M, McCormick S, Xing Z. Immunopathology in influenza virus infection: Uncoupling the friend from foe. Clin Immunol 2012; 144:57-69. [DOI: 10.1016/j.clim.2012.05.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 04/30/2012] [Accepted: 05/08/2012] [Indexed: 12/23/2022]
|
44
|
St Paul M, Mallick AI, Read LR, Villanueva AI, Parvizi P, Abdul-Careem MF, Nagy É, Sharif S. Prophylactic treatment with Toll-like receptor ligands enhances host immunity to avian influenza virus in chickens. Vaccine 2012; 30:4524-31. [PMID: 22531557 DOI: 10.1016/j.vaccine.2012.04.033] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 04/06/2012] [Accepted: 04/09/2012] [Indexed: 12/18/2022]
Abstract
Avian influenza viruses (AIV) pose a threat towards the health of both poultry and humans. To interrupt the transmission of the virus, novel prophylactic strategies must be considered which may reduce the shedding of AIV. One potential is the prophylactic use of Toll-like receptor (TLR) ligands. Many cells of the immune system express TLRs, and cellular responses to TLR stimulation include activation and the production of cytokines. TLR ligands have been employed as prophylactic treatments to enhance host resistance to pathogens both in mammals and chickens. Therefore, the present study was conducted to determine whether TLR ligands may be used prophylactically in chickens to enhance host immunity to AIV. Chickens received intramuscular injections of either low or high doses of the TLR ligands poly I:C, lipopolysaccharide (LPS) and CpG ODN. Twenty-four hours post-treatment, chickens were infected with the low pathogenic avian influenza virus H4N6, and both oropharyngeal and cloacal virus shedding were assessed on days 4 and 7 post-infection. To identify potential correlates of immunity, spleen and lungs were collected on days 2, 4 and 7 post-infection for RNA extraction. The results suggested that all of the TLR ligand treatments induced a significant reduction in virus shedding, with the TLR3 ligand poly I:C conferring the greatest AIV immunity compared to control birds, followed by CpG ODN and LPS. Furthermore, transcriptional analysis of gene expression in the spleen and lungs suggest IFN-α and IL-8 as correlates of immunity conferred by poly I:C, and IFN-γ for CpG ODN and LPS. In conclusion, TLR ligands, have the ability to enhance host immunity against AIV, and future studies should consider exploring the combinatory effects of poly I:C and CpG ODN prophylaxis in conjunction with AIV vaccination.
Collapse
Affiliation(s)
- Michael St Paul
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | | | | | | | | | | | | | | |
Collapse
|