1
|
Santamarina-Fernández R, Fuentes-Valverde V, Silva-Rodríguez A, García P, Moscoso M, Bou G. Pseudomonas aeruginosa Vaccine Development: Lessons, Challenges, and Future Innovations. Int J Mol Sci 2025; 26:2012. [PMID: 40076637 PMCID: PMC11900337 DOI: 10.3390/ijms26052012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen with a multidrug-resistant profile that has become a critical threat to global public health. It is one of the main causes of severe nosocomial infections, including ventilator-associated pneumonia, chronic infections in patients with cystic fibrosis, and bloodstream infections in immunosuppressed individuals. Development of vaccines against P. aeruginosa is a major challenge owing to the high capacity of this bacterium to form biofilms, its wide arsenal of virulence factors (including secretion systems, lipopolysaccharides, and outer membrane proteins), and its ability to evade the host immune system. This review provides a comprehensive historical overview of vaccine development efforts targeting this pathogen, ranging from early attempts in the 1970s to recent advancements, including vaccines based on novel proteins and emerging technologies such as nanoparticles and synthetic conjugates. Despite numerous promising preclinical developments, very few candidates have progressed to clinical trials, and none have achieved final approval. This panorama highlights the significant scientific efforts undertaken and the inherent complexity of successfully developing an effective vaccine against P. aeruginosa.
Collapse
Affiliation(s)
- Rebeca Santamarina-Fernández
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
| | - Víctor Fuentes-Valverde
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Área de Medicamentos Biológicos, Agencia Española de Medicamentos y Productos Sanitarios (AEMPS), 28022 Madrid, Spain
| | - Alis Silva-Rodríguez
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
| | - Patricia García
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miriam Moscoso
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Germán Bou
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, 15006 A Coruña, Spain; (R.S.-F.); (V.F.-V.); (A.S.-R.); (P.G.); (G.B.)
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Universidad de A Coruña, 15006 A Coruña, Spain
| |
Collapse
|
2
|
Korpi F, Irajian G, Forouhi F, Mohammadian T. A chimeric vaccine targeting Pseudomonas aeruginosa virulence factors protects mice against lethal infection. Microb Pathog 2023; 178:106033. [PMID: 36813005 DOI: 10.1016/j.micpath.2023.106033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/25/2023] [Accepted: 02/12/2023] [Indexed: 02/22/2023]
Abstract
Pseudomonas aeruginosa is an important and hazardous nosocomial pathogen in respiratory tract infections and rapidly achieves antibiotic resistance, so it is necessary to develop an effective vaccine to combat the infection. The Type III secretion system (T3SS) protein P. aeruginosa V-antigen (PcrV), outer membrane protein F (OprF), and two kinds of flagellins (FlaA and FlaB) all play important roles in the pathogenesis of P. aeruginosa lung infection and its spread into deeper tissues. In a mouse acute pneumonia model, the protective effects of a chimer vaccine including PcrV, FlaA, FlaB, and OprF (PABF) protein were investigated. PABF immunization prompted robust opsonophagocytic titer of IgG antibodies and decreased bacterial burden, and improved survival afterward intranasal challenge with ten times 50% lethal doses (LD50) of P. aeruginosa strains, indicating its broad-spectrum immunity. Moreover, these findings showed a promise chimeric vaccine candidate to treat and control P. aeruginosa infections.
Collapse
Affiliation(s)
- Fatemeh Korpi
- Department of Cell and Molecular Biology, Faculty of Basic Science, Islamic Azad University Shahre Qods Branch, Iran
| | - Gholamreza Irajian
- Department of Cell and Molecular Biology, Faculty of Basic Science, Islamic Azad University Shahre Qods Branch, Iran; Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Forouhi
- Department of Cell and Molecular Biology, Faculty of Basic Science, Islamic Azad University Shahre Qods Branch, Iran
| | - Taher Mohammadian
- Department of Cell and Molecular Biology, Faculty of Basic Science, Islamic Azad University Shahre Qods Branch, Iran
| |
Collapse
|
3
|
Iraji D, Oftedal BE, Wolff ASB. Th17 Cells: Orchestrators of Mucosal Inflammation and Potential Therapeutic Targets. Crit Rev Immunol 2023; 43:25-52. [PMID: 37831521 DOI: 10.1615/critrevimmunol.2023050360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
T helper 17 (Th17) cells represent a specialized subgroup of effector CD4+ T cells known for their role in provoking neutrophil-driven tissue inflammation, particularly within mucosal tissues. Although they are pivotal for defending the host against extracellular bacteria and fungi, they have also been associated with development of various T cell-mediated inflammatory conditions, autoimmune diseases, and even cancer. Notably, Th17 cells exhibit a dual nature, with different Th17 cell subtypes showcasing distinct effector functions and varying capacities to incite autoimmune tissue inflammation. Furthermore, Th17 cells exhibit significant plasticity, which carries important functional implications, both in terms of their expression of cytokines typically associated with other effector T cell subsets and in their interactions with regulatory CD4+ T cells. The intricate balance of Th17 cytokines can also be a double-edged sword in inflammation, autoimmunity, and cancer. Within this article, we delve into the mechanisms that govern the differentiation, function, and adaptability of Th17 cells. We culminate with an exploration of therapeutic potentials in harnessing the power of Th17 cells and their cytokines. Targeted interventions to modulate Th17 responses are emerging as promising strategies for autoimmunity, inflammation, and cancer treatment. By precisely fine-tuning Th17-related pathways, we may unlock new avenues for personalized therapeutic approaches, aiming to restore immune balance, alleviate the challenges of these disorders, and ultimately enhance the quality of life for individuals affected by them.
Collapse
Affiliation(s)
- Dorsa Iraji
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bergithe E Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
4
|
Hamad AS, Edward EA, Sheta E, Aboushleib HM, Bahey-El-Din M. Iron Acquisition Proteins of Pseudomonas aeruginosa as Potential Vaccine Targets: In Silico Analysis and In Vivo Evaluation of Protective Efficacy of the Hemophore HasAp. Vaccines (Basel) 2022; 11:vaccines11010028. [PMID: 36679873 PMCID: PMC9864456 DOI: 10.3390/vaccines11010028] [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: 11/09/2022] [Revised: 12/12/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Pseudomonas aeruginosa (PA) is a Gram-negative pathogen responsible for fatal nosocomial infections worldwide. Iron is essential for Gram-negative bacteria to establish an infection. Therefore, iron acquisition proteins (IAPs) of bacteria are attractive vaccine targets. METHODOLOGY A "Reverse Vaccinology" approach was employed in the current study. Expression levels of 37 IAPs in various types of PA infections were analyzed in seven previously published studies. The IAP vaccine candidate was selected based on multiple criteria, including a high level of expression, high antigenicity, solubility, and conservation among PA strains, utilizing suitable bioinformatics analysis tools. The selected IAP candidate was recombinantly expressed in Escherichia coli and purified using metal affinity chromatography. It was further evaluated in vivo for protection efficacy. The novel immune adjuvant, naloxone (NAL), was used. RESULTS AND DISCUSSION HasAp antigen met all the in silico selection criteria, being highly antigenic, soluble, and conserved. In addition, it was the most highly expressed IAP in terms of average fold change compared to control. Although HasAp did excel in the in silico evaluation, subcutaneous immunization with recombinant HasAp alone or recombinant HasAp plus NAL (HasAP-NAL) did not provide the expected protection compared to controls. Immunized mice showed a low IgG2a/IgG1 ratio, indicating a T-helper type 2 (Th2)-oriented immune response that is suboptimal for protection against PA infections. Surprisingly, the bacterial count in livers of both NAL- and HasAp-NAL-immunized mice was significantly lower than the count in the HasAp and saline groups. The same trend was observed in kidneys and lungs obtained from these groups, although the difference was not significant. Such protection could be attributed to the enhancement of innate immunity by NAL. CONCLUSIONS We provided a detailed in silico analysis of IAPs of PA followed by in vivo evaluation of the best IAP, HasAp. Despite the promising in silico results, HasAp did not provide the anticipated vaccine efficacy. HasAp should be further evaluated as a vaccine candidate through varying the immunization regimens, models of infection, and immunoadjuvants. Combination with other IAPs might also improve vaccination efficacy. We also shed light on several highly expressed promising IAPs whose efficacy as vaccine candidates is worthy of further investigation.
Collapse
Affiliation(s)
- Abdelrahman S. Hamad
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria P.O. Box 25435, Egypt
| | - Eva A. Edward
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria P.O. Box 25435, Egypt
| | - Eman Sheta
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria P.O. Box 21131, Egypt
| | - Hamida M. Aboushleib
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria P.O. Box 25435, Egypt
| | - Mohammed Bahey-El-Din
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria P.O. Box 25435, Egypt
- Correspondence:
| |
Collapse
|
5
|
Multicomponent Pseudomonas aeruginosa Vaccines Eliciting Th17 Cells and Functional Antibody Responses Confer Enhanced Protection against Experimental Acute Pneumonia in Mice. Infect Immun 2022; 90:e0020322. [PMID: 36069593 PMCID: PMC9584304 DOI: 10.1128/iai.00203-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The Gram-negative pathogen Pseudomonas aeruginosa is a common cause of pneumonia in hospitalized patients. Its increasing antibiotic resistance and widespread occurrence present a pressing need for vaccines. We previously showed that a P. aeruginosa type III secretion system protein, PopB, elicits a strong Th17 response in mice after intranasal (IN) immunization and confers antibody-independent protection against pneumonia in mice. In the current study, we evaluated the immunogenicity and protective efficacy in mice of the combination of PopB (purified with its chaperone protein PcrH) and OprF/I, an outer membrane hybrid fusion protein, compared with immunization with the proteins individually either by the intranasal (IN) or subcutaneous (SC) routes. Our results show that after vaccination, a Th17 recall response from splenocytes was detected only in mice vaccinated with PopB/PcrH, either alone or in combination with OprF/I. Mice immunized with the combination of PopB/PcrH and OprF/I had enhanced protection in an acute lethal P. aeruginosa pneumonia model, regardless of vaccine route, compared with mice vaccinated with either alone or adjuvant control. Immunization generated IgG titers against the vaccine proteins and whole P. aeruginosa cells. Interestingly, none of these antisera had opsonophagocytic killing activity, but antisera from mice immunized with vaccines containing OprF/I, had the ability to block IFN-γ binding to OprF/I, a known virulence mechanism. Hence, vaccines combining PopB/PcrH with OprF/I that elicit functional antibodies lead to a broadly and potently protective vaccine against P. aeruginosa pulmonary infections.
Collapse
|
6
|
Pemphigus during the COVID-19 Epidemic: Infection Risk, Vaccine Responses and Management Strategies. J Clin Med 2022; 11:jcm11143968. [PMID: 35887732 PMCID: PMC9317200 DOI: 10.3390/jcm11143968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/23/2022] [Accepted: 07/06/2022] [Indexed: 01/08/2023] Open
Abstract
Pemphigus is a rare autoimmune blistering disease, involving potentially life-threatening conditions often requiring immunosuppression. Currently, the COVID-19 pandemic caused by severe acute respiratory disease coronavirus 2 (SARS-CoV-2) infection has become a global public emergency. Vaccines are the most effective defense against COVID-19 infection. However, in clinic, there are cases of new onset or flare of pemphigus following COVID-19 vaccination, where vaccines have manifested significantly desirable risk-benefit profiles for patients. Although Rituximab, as first-line therapy, may impair humoral immunity, pemphigus may not predispose to develop COVID-19 infection compared to a healthy population. Conversely, delay or interruption of immunosuppressants probably results in unfavorable clinical outcomes for disease progression. Overall, clinicians should encourage their patients to undergo the vaccination after a comprehensive assessment. The definite association between COVID-19 vaccination and pemphigus remains to be further elucidated. Herein, we provide an overview of the published studies to date on COVID-19 and pemphigus as well as the exploration of their complicated interplay. In addition, we discuss the management strategies for pemphigus patients in this special period, in an effort to more effectively establish a standard treatment paradigm for this particular patient group.
Collapse
|
7
|
Galantini MPL, Leal LS, Rodrigues KB, Ribeiro IS, Pereira IS, Gonçalves CV, Calado SPM, Santos DPD, Muniz IPR, Silva RAADA. Physical activity reduces intradermal bacterial load in a murine model submitted to forced swim training - a pilot study. AN ACAD BRAS CIENC 2022; 94:e20200561. [PMID: 35703689 DOI: 10.1590/0001-3765202220200561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/15/2020] [Indexed: 11/22/2022] Open
Abstract
Regular exercise is beneficial to health. This study evaluated the effects of moderate and intense physical exercise modalities on intradermal infection by Staphylococcus aureus in a murine model. Mice that practiced moderate exercise had lower bacterial load on lymph nodes and less inflammatory infiltrate in dermis. They presented greater weight, however, less amount of epididymal fat: the weight was increased while they had fat diminished. A positive correlation was observed between lipid content and bacterial load in mice trained at moderate intensity. Animals that were under high intensity exercises presented superior bacterial load on the lymph nodes, increased neutrophil count and circulating lymphocytes, and had leukocyte recruitment to the dermis augmented, when compared to the ones in moderate exercise. These findings suggest that moderate physical activity modulates the immune response in dermal infection caused by S. aureus in a murine model.
Collapse
Affiliation(s)
- Maria P L Galantini
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| | - Lorena S Leal
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| | - Karine B Rodrigues
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| | - Israel S Ribeiro
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| | - Italo S Pereira
- Universidade de São Paulo (USP), Faculdade de Medicina de Ribeirão Preto (FMRP), Departamento de Bioquímica e Imunologia, Avenida Bandeirantes, 3900, 14049-900 Ribeirão Preto, SP, Brazil
| | - Caroline V Gonçalves
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| | - Stéfano P M Calado
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| | - Denisar P Dos Santos
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| | - Igor P R Muniz
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| | - Robson A A DA Silva
- Universidade Federal da Bahia (UFBA), Instituto Multidisciplinar em Saúde (IMS), Campus Anísio Teixeira (CAT), Rua Hormindo Barros, 58, 45029-094 Vitória da Conquista, BA, Brazil
| |
Collapse
|
8
|
Treatment of Moderate to Severe Psoriasis during the COVID-19 Pandemic: Lessons Learned and Opportunities. J Clin Med 2022; 11:jcm11092422. [PMID: 35566548 PMCID: PMC9101352 DOI: 10.3390/jcm11092422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 01/27/2023] Open
Abstract
Since the beginning of the coronavirus disease 2019 (COVID-19) pandemic, clinicians have been overwhelmed by questions beyond the SARS-CoV-2 infection itself. In dermatology practice, clinicians have been facing difficulties concerning therapeutic management of chronic immune-mediated skin disease, above all psoriasis. Major challenges arisen were to understand the role of immunosuppression or immunomodulation on COVID-19 evolution, the benefit/risk ratio related to discontinuation or modification of ongoing treatment, and the appropriateness of initiating new treatments, the optimization of timing in vaccination administration to patients under immunomodulatory treatments, and finally how to find new strategy of patients’ management through remote assistance. In this comprehensive review, we present the current evidence about the course and management of psoriasis during the COVID-19 pandemic. The general message from dermatologists was that data did not suggest that having PSO or its treatment significantly increased risk of SARS-CoV-2 infection or more severe COVID-19 course, the vaccination is highly recommended in all psoriatic patients, beyond ongoing treatment, and that the telehealth experience was a success overall.
Collapse
|
9
|
Gonzaga ZJC, Zhang J, Rehm BHA. Intranasal Delivery of Antigen-Coated Polymer Particles Protects against Pseudomonas aeruginosa Infection. ACS Infect Dis 2022; 8:744-756. [PMID: 35238554 DOI: 10.1021/acsinfecdis.1c00434] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen that is intrinsically resistant to multiple antibiotics, causing severe and persistent infections in immunocompromised individuals. This bacterium has been listed as a priority pathogen by the WHO in 2017, and there is no vaccine available for human use. In this study, 10 vaccine candidate antigens were selected for particulate vaccine design. We engineered Escherichia coli to assemble biopolymer particles (BPs) that were either coated with epitopes (Ag) derived from OprF/I-AlgE proteins or PopB or PopB-Ag or coated with single or double copies of epitopes (10Ag and 10Ag(2x)) derived from OprF, OprI, AlgE, OprL, PopB, PilA, PilO, FliC, Hcp1, and CdrA. Antigen-coated BPs showed a diameter of 0.93-1.16 μm with negative surface charge. Antigens attached to BPs were identified by mass spectrometry. Vaccination with BP-Ag, BP-PopB, BP-PopBAg, PB-10Ag, and BP-10Ag(2x) with and without Alhydrogel adjuvant induced significant antigen-specific humoral and cell-mediated immune responses in mice. All particulate vaccines with Alhydrogel induced protection in an acute pneumonia murine model of P. aeruginosa infection, contributing to up to 80% survival when administered intramuscularly, and the addition of Alhydrogel boosted immunity. The BP-10Ag(2x) vaccine candidate showed the best performance and even induced protective immunity in the absence of Alhydrogel. Intramuscular administration of the BP-10Ag(2x) without Alhydrogel vaccine resulted in 60% survival. Intranasal vaccination induced immunity, contributing to about 90% survival. Overall, our data suggest that vaccination with BPs coated with P. aeruginosa antigens induce protective immunity against P. aeruginosa infections. The possibility of intranasal delivery will strongly facilitate administration and use of BP vaccines.
Collapse
Affiliation(s)
- Zennia Jean C. Gonzaga
- Centre for Cell Factories and Biopolymers (CCFB), Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
| | - Jinyong Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing 400038, PR China
| | - Bernd H. A. Rehm
- Centre for Cell Factories and Biopolymers (CCFB), Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, Queensland 4111, Australia
- Menzies Health Institute Queensland (MHIQ), Griffith University, Gold Coast, Queensland 4222, Australia
| |
Collapse
|
10
|
Outer Membrane Vesicles Displaying a Heterologous PcrV-HitA Fusion Antigen Promote Protection against Pulmonary Pseudomonas aeruginosa Infection. mSphere 2021; 6:e0069921. [PMID: 34612675 PMCID: PMC8510544 DOI: 10.1128/msphere.00699-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Along with surging threats and antibiotic resistance of Pseudomonas aeruginosa in health care settings, it is imperative to develop effective vaccines against P. aeruginosa infection. In this study, we used an Asd (aspartate-semialdehyde dehydrogenase)-based balanced-lethal host-vector system of a recombinant Yersinia pseudotuberculosis mutant to produce self-adjuvanting outer membrane vesicles (OMVs). The OMVs were used as a carrier to deliver the heterologous PcrV-HitAT (PH) fusion antigen of P. aeruginosa for vaccine evaluation. Intramuscular vaccination with OMVs carrying the PH antigen (referred to rOMV-PH) afforded 73% protection against intranasal challenge with 5 × 106 (25 50% lethal doses) of the cytotoxic PA103 strain and complete protection against a noncytotoxic PAO1 strain. In contrast, vaccination with the PH-deficient OMVs or PH antigen alone failed to offer effective protection against the same challenge. Immune analysis showed that the rOMV-PH vaccination induced potent humoral and Th1/Th17 responses compared to the PH vaccination. The rOMV-PH vaccination rapidly cleared P. aeruginosa burdens with coordinated production of proinflammatory cytokines in mice. Moreover, antigen-specific CD4+ and CD8+ T cells and their producing cytokines (tumor necrosis factor alpha and interleukin-17A), rather than antibodies, were essential for protection against pneumonic P. aeruginosa infection. Our studies demonstrated that the recombinant Y. pseudotuberculosis OMVs delivering heterologous P. aeruginosa antigens could be a new promising vaccine candidate for preventing the spread of drug-resistant P. aeruginosa. IMPORTANCE Hospital- and community-acquired infections with Pseudomonas aeruginosa cause a high rate of morbidity and mortality in patients who have underlying medical conditions. The spread of multidrug-resistant P. aeruginosa strains is becoming a great challenge for treatment using antibiotics. Thus, a vaccine as one of the alternative strategies is urgently required to prevent P. aeruginosa infection.
Collapse
|
11
|
Diotallevi F, Campanati A, Radi G, Martina E, Rizzetto G, Barbadoro P, D'Errico MM, Offidani A. Vaccines Against SARS-CoV-2 in Psoriasis Patients on Immunosuppressive Therapy: Implications of Vaccination Nationwide Campaign on Clinical Practice in Italy. Dermatol Ther (Heidelb) 2021; 11:1889-1903. [PMID: 34586598 PMCID: PMC8480269 DOI: 10.1007/s13555-021-00610-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Indexed: 01/08/2023] Open
Abstract
More than 12 months have passed since the World Health Organization (WHO) declared Coronavirus Disease 19 (COVID-19), caused by the SARS-CoV2 virus, to be a pandemic on 11 March 2020. The entire world scientific community agrees that at this time vaccine is the most promising weapon to combat the infection and the severity of the disease. According to the document “Draft landscape of COVID-19 candidate vaccines” by WHO, 272 vaccines against SARS-CoV-2 virus are in development, although only four of these, produced by Pfizer-BioNTech (Pfizer, Inc. and BioNTech), Moderna, AstraZeneca, and Janssen companies, respectively, have been approved by European Medicines Agency and Italian Medicines Agency and subsequently distributed nationwide for use. These vaccines are the result of highly innovative procedures and are quite different from each other in terms of composition. Even clinicians in various medical fields may be unfamiliar with the effects of these vaccines. There is the strong emerging need for dermatologists to understand the crucial role of vaccines, with a focus on the need to vaccinate patients suffering from immune-mediated skin diseases, such as psoriasis, while taking the ongoing treatment into consideration regarding the timing of vaccination. Similarly, psoriasis patients aware of having an immune-mediated and inflammatory disease are increasingly asking the dermatologist information about the efficacy and safety of vaccines against SARS-CoV-2 virus. In this narrative review of the literature and critical analysis of the recommendations of the Italian Ministry of Health, we analyze the implications of the vaccination campaign on dermatological patients with psoriasis undergoing immunosuppressive treatment.
Collapse
Affiliation(s)
- Federico Diotallevi
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| | - Anna Campanati
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| | - Giulia Radi
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy.
| | - Emanuela Martina
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| | - Giulio Rizzetto
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| | - Pamela Barbadoro
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, Ancona, Italy
| | - Marcello Mario D'Errico
- Department of Biomedical Sciences and Public Health, Section of Hygiene, Preventive Medicine and Public Health, Polytechnic University of the Marche Region, Ancona, Italy
| | - Annamaria Offidani
- Department of Clinical and Molecular Sciences, Dermatological Clinic, Polytechnic University of the Marche Region, Via Conca 71, 60020, Ancona, Italy
| |
Collapse
|
12
|
Bostan E, Elmas L, Yel B, Yalici-Armagan B. Exacerbation of plaque psoriasis after inactivated and BNT162b2 mRNA COVID-19 vaccines: A report of two cases. Dermatol Ther 2021; 34:e15110. [PMID: 34427024 PMCID: PMC8646432 DOI: 10.1111/dth.15110] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 08/22/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Ecem Bostan
- Department of Dermatology and Venereology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Leyla Elmas
- Department of Dermatology and Venereology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Beril Yel
- Department of Dermatology and Venereology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Basak Yalici-Armagan
- Department of Dermatology and Venereology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
13
|
Recombinant Pseudomonas bio-nanoparticles induce protection against pneumonic Pseudomonas aeruginosa infection. Infect Immun 2021; 89:e0039621. [PMID: 34310892 DOI: 10.1128/iai.00396-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To develop an effective Pseudomonas aeruginosa (PA) outer-membrane-vesicles (OMVs) vaccine, we eliminated multiple virulence factors from a wild-type P. aeruginosa PA103 strain (PA103) to generate a recombinant strain, PA-m14. The PA-m14 strain was tailored with a pSMV83 plasmid encoding the pcrV-hitAT fusion gene to produce OMVs. The recombinant OMVs enclosed increased amounts of PcrV-HitAT bivalent antigen (PH) (termed OMV-PH) and exhibited reduced toxicity compared to the OMVs from PA103. Intramuscular vaccination with OMV-PH from PA-m14(pSMV83) afforded 70% protection against intranasal challenge with 6.5 × 106 CFU (∼30 LD50) of PA103, while immunization using OMVs without the PH antigen (termed OMV-NA) or the PH antigen alone failed to offer effective protection against the same challenge. Further immune analysis showed that the OMV-PH immunization significantly stimulated potent antigen-specific humoral and T-cell (Th1/Th17) responses in comparison to the PH or OMV-NA immunization in mice, which can effectively hinder PA infection. Undiluted anti-sera from OMV-PH-immunized mice displayed significant opsonophagocytic killing of WT PA103 compared to antisera from PH antigen- or OMV-NA-immunized mice. Moreover, the OMV-PH immunization afforded significant antibody-indentpednet cross-protection to mice against PAO1 and a clinical isolate AMC-PA10 strains. Collectively, the recombinant PA OMV delivering the PH bivalent antigen exhibits high immunogenicity and would be a promising next-generation vaccine candidate against PA infection.
Collapse
|
14
|
Gonzaga ZJC, Merakou C, DiGiandomenico A, Priebe GP, Rehm BHA. A Pseudomonas aeruginosa-Derived Particulate Vaccine Protects against P. aeruginosa Infection. Vaccines (Basel) 2021; 9:803. [PMID: 34358220 PMCID: PMC8309987 DOI: 10.3390/vaccines9070803] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 01/05/2023] Open
Abstract
Despite numerous efforts to develop an effective vaccine against Pseudomonas aeruginosa, no vaccine has yet been approved for human use. This study investigates the utility of the P. aeruginosa inherently produced polyhydroxyalkanaote (PHA) inclusions and associated host-cell proteins (HCP) as a particulate vaccine platform. We further engineered PHA inclusions to display epitopes derived from the outer membrane proteins OprF/OprI/AlgE (Ag) or the type III secretion system translocator PopB. PHA and engineered PHA beads induced antigen-specific humoral, cell-mediated immune responses, anti-HCP and anti-polysaccharide Psl responses in mice. Antibodies mediated opsonophagocytic killing and serotype-independent protective immunity as shown by 100% survival upon challenge with P. aeruginosa in an acute pneumonia murine model. Vaccines were stable at 4 °C for at least one year. Overall, our data suggest that vaccination with subcellular empty PHA beads was sufficient to elicit multiple immune effectors that can prevent P. aeruginosa infection.
Collapse
Affiliation(s)
- Zennia Jean C. Gonzaga
- Centre for Cell Factories and Biopolymers (CCFB), Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, QLD 4111, Australia;
| | - Christina Merakou
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA; (C.M.); (G.P.P.)
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
| | - Antonio DiGiandomenico
- Discovery Microbiome, Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 34321, USA;
| | - Gregory P. Priebe
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA 02115, USA; (C.M.); (G.P.P.)
- Department of Anaesthesia, Harvard Medical School, Boston, MA 02115, USA
- Division of Infectious Diseases, Department of Pediatrics, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Bernd H. A. Rehm
- Centre for Cell Factories and Biopolymers (CCFB), Griffith Institute for Drug Discovery, Griffith University, Don Young Road, Nathan, QLD 4111, Australia;
- Menzies Health Institute Queensland (MHIQ), Griffith University, Gold Coast, QLD 4222, Australia
| |
Collapse
|
15
|
Solimani F, Mansour Y, Didona D, Dilling A, Ghoreschi K, Meier K. Development of severe pemphigus vulgaris following SARS-CoV-2 vaccination with BNT162b2. J Eur Acad Dermatol Venereol 2021; 35:e649-e651. [PMID: 34169588 PMCID: PMC8447452 DOI: 10.1111/jdv.17480] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/23/2021] [Indexed: 12/20/2022]
Affiliation(s)
- F Solimani
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Y Mansour
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - D Didona
- Department of Dermatology and Allergology, Philipps University, Marburg, Germany
| | - A Dilling
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - K Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - K Meier
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
16
|
Zhu Y, Xu Y, Hong L, Zhou C, Chen J. Immunization With a DNA Vaccine Encoding the Toxoplasma gondii' s GRA39 Prolongs Survival and Reduce Brain Cyst Formation in a Murine Model. Front Microbiol 2021; 12:630682. [PMID: 33995293 PMCID: PMC8113873 DOI: 10.3389/fmicb.2021.630682] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/29/2021] [Indexed: 12/02/2022] Open
Abstract
Toxoplasma gondii, an obligate intracellular protozoan parasite, can cause infect almost all warm-blooded animals and humans. To evaluate the immunogenicity and protective efficacy of T. gondii GRA39 (TgGRA39) in mice by using DNA immunization, we constructed a recombinant eukaryotic plasmid pVAX-TgGRA39. The specific immune responses in immunized mice were analyzed by serum antibody and cytokine measurements, lymphocyte proliferation assays and flow cytometry of T lymphocyte subclasses. Also, protective efficacy against acute and chronic T. gondii infection was assessed by observing the survival time after challenge with the highly virulent T. gondii RH strain (Genotype I) and counting the number of cyst-forming in brain at 4 weeks post-infection with the cyst-forming PRU strain of T. gondii (Genotype II), respectively. Our results showed that DNA immunization with pVAX-GRA39 via intramuscular injection three times, at 2-week intervals could elicit humoral and cellular immune response, indicated by enhanced levels of IgG and IgG2a antibodies (a slightly elevated IgG2a to IgG1 ratio), and increased levels of cytokines IFN-γ, IL-2, IL-12, IL-17A, IL-17F, IL-22 and IL-23 and percentages of CD3+ CD4+ CD8- and CD3+ CD8+ CD4– T cells, in contrast to non-immunized mice. The significant increase in the expression levels of IL-6, TGF-β1, IL-1β, and the transcription factor factors RORγt, RORα, and STAT3 involved in the activation and pathway of Th17 and Tc17 cells, were also observed. However, no significant difference was detected in level of IL-4 and IL-10 (p > 0.05). These effective immune responses had mounted protective immunity against T. gondii infection, with a prolonged survival time (16.80 ± 3.50 days) and reduced cyst numbers (44.5%) in comparison to the control mice. Our data indicated that pVAX-TgGRA39 could induce effective humoral, and Th1-type, Th17, and Tc17 cellular immune responses, and may represent a promising vaccine candidate against both acute and chronic T. gondii infection.
Collapse
Affiliation(s)
- Yuchao Zhu
- Department of Radiology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Yanan Xu
- The Ningbo Women and Children's Hospital, Ningbo, China
| | - Lu Hong
- Department of Radiology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China
| | - Chunxue Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jia Chen
- Department of Radiology, The Affiliated Hospital of Medical School of Ningbo University, Ningbo, China.,The Ningbo Women and Children's Hospital, Ningbo, China
| |
Collapse
|
17
|
Ma C, Chen W. Where are we and how far is there to go in the development of an Acinetobacter vaccine? Expert Rev Vaccines 2021; 20:281-295. [PMID: 33554671 DOI: 10.1080/14760584.2021.1887735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Healthcare-associated infections caused by multidrug-resistant Acinetobacter baumannii are becoming alarming worldwide. However, the pipeline of new antibiotics is very limited. Vaccination is one of the most cost effective and promising strategies to prevent infections and can play an important role in combat multidrug resistance A. baumannii and prevent the development of new drug resistance. AREA COVERED This review gives an overview of the research and development of A. baumannii vaccines during the past five years (2015-2020), discusses the key progresses and current challenges of the field, and speculates on the future of A. baumannii vaccine development. EXPERT OPINION Moderate progresses have been made in the research and development of A. baumannii vaccine in the last five years, in particular in the areas of identification of new protein targets, development of multicomponent vaccines, and use of vaccines and antibodies as adjuncts for antibiotics therapies. However, substantial scientific and logistic challenges, such as selection of lead vaccine candidates and formulation, vaccine clinical trials and targeted population, and financial incentives, remain. Thus, innovative strategies will be needed before an A. baumannii vaccine candidate can be brought into late stage of preclinical development in next five years.
Collapse
Affiliation(s)
- Crystal Ma
- Human Health Therapeutics Research Center (HHT), National Research Council Canada, Ottawa, Ontario Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center (HHT), National Research Council Canada, Ottawa, Ontario Canada.,Department of Biology, Brock University, St. Catharines, Ontario Canada
| |
Collapse
|
18
|
Wang Y, Cheng X, Wan C, Wei J, Gao C, Zhang Y, Zeng H, Peng L, Luo P, Lu D, Zou Q, Gu J. Development of a Chimeric Vaccine Against Pseudomonas aeruginosa Based on the Th17-Stimulating Epitopes of PcrV and AmpC. Front Immunol 2021; 11:601601. [PMID: 33552056 PMCID: PMC7859429 DOI: 10.3389/fimmu.2020.601601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/03/2020] [Indexed: 11/16/2022] Open
Abstract
Pulmonary infection caused by Pseudomonas aeruginosa (PA) has created an urgent need for an efficient vaccine, but the protection induced by current candidates is limited, partially because of the high variability of the PA genome. Antigens targeting pulmonary Th17 responses are able to provide antibody-independent and broad-spectrum protection; however, little information about Th17-stimulating antigens in PA is available. Herein, we identified two novel PA antigens that effectively induce Th17-dependent protection, namely, PcrV (PA1706) and AmpC (PA4110). Compared to intramuscular immunization, intranasal immunization enhanced the protection of rePcrV due to activation of a Th17 response. The Th17-stimulating epitopes of PcrV and AmpC were identified, and the recombinant protein PVAC was designed and generated by combining these Th17-stimulating epitopes. PVAC was successfully produced in soluble form and elicited broad protective immunity against PA. Our results provide an alternative strategy for the development of Th17-based vaccines against PA and other pathogens.
Collapse
Affiliation(s)
- Ying Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xin Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Chuang Wan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jinning Wei
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Chen Gao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yi Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Liusheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jiang Gu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
19
|
IL-17 mediates protective immunity against nasal infection with Bordetella pertussis by mobilizing neutrophils, especially Siglec-F + neutrophils. Mucosal Immunol 2021; 14:1183-1202. [PMID: 33976385 PMCID: PMC8379078 DOI: 10.1038/s41385-021-00407-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 02/04/2023]
Abstract
Understanding the mechanism of protective immunity in the nasal mucosae is central to the design of more effective vaccines that prevent nasal infection and transmission of Bordetella pertussis. We found significant infiltration of IL-17-secreting CD4+ tissue-resident memory T (TRM) cells and Siglec-F+ neutrophils into the nasal tissue during primary infection with B. pertussis. Il17A-/- mice had significantly higher bacterial load in the nasal mucosae, associated with significantly reduced infiltration of Siglec-F+ neutrophils. Re-infected convalescent mice rapidly cleared B. pertussis from the nasal cavity and this was associated with local expansion of IL-17-producing CD4+ TRM cells. Depletion of CD4 T cells from the nasal tissue during primary infection or after re-challenge of convalescent mice significantly delayed clearance of bacteria from the nasal mucosae. Protection was lost in Il17A-/- mice and this was associated with significantly less infiltration of Siglec-F+ neutrophils and antimicrobial peptide (AMP) production. Finally, depletion of neutrophils reduced the clearance of B. pertussis following re-challenge of convalescent mice. Our findings demonstrate that IL-17 plays a critical role in natural and acquired immunity to B. pertussis in the nasal mucosae and this effect is mediated by mobilizing neutrophils, especially Siglec-F+ neutrophils, which have high neutrophil extracellular trap (NET) activity.
Collapse
|
20
|
Sainz-Mejías M, Jurado-Martín I, McClean S. Understanding Pseudomonas aeruginosa-Host Interactions: The Ongoing Quest for an Efficacious Vaccine. Cells 2020; 9:cells9122617. [PMID: 33291484 PMCID: PMC7762141 DOI: 10.3390/cells9122617] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/18/2022] Open
Abstract
Pseudomonas aeruginosa is a leading cause of chronic respiratory infections in people with cystic fibrosis (CF), bronchiectasis or chronic obstructive pulmonary disease (COPD), and acute infections in immunocompromised individuals. The adaptability of this opportunistic pathogen has hampered the development of antimicrobial therapies, and consequently, it remains a major threat to public health. Due to its antimicrobial resistance, vaccines represent an alternative strategy to tackle the pathogen, yet despite over 50 years of research on anti-Pseudomonas vaccines, no vaccine has been licensed. Nevertheless, there have been many advances in this field, including a better understanding of the host immune response and the biology of P. aeruginosa. Multiple antigens and adjuvants have been investigated with varying results. Although the most effective protective response remains to be established, it is clear that a polarised Th2 response is sub-optimal, and a mixed Th1/Th2 or Th1/Th17 response appears beneficial. This comprehensive review collates the current understanding of the complexities of P. aeruginosa-host interactions and its implication in vaccine design, with a view to understanding the current state of Pseudomonal vaccine development and the direction of future efforts. It highlights the importance of the incorporation of appropriate adjuvants to the protective antigen to yield optimal protection.
Collapse
|
21
|
Gupta G, Mou Z, Jia P, Sharma R, Zayats R, Viana SM, Shan L, Barral A, Boaventura VS, Murooka TT, Soussi-Gounni A, de Oliveira CI, Uzonna JE. The Long Pentraxin 3 (PTX3) Suppresses Immunity to Cutaneous Leishmaniasis by Regulating CD4 + T Helper Cell Response. Cell Rep 2020; 33:108513. [PMID: 33326783 DOI: 10.1016/j.celrep.2020.108513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 06/30/2020] [Accepted: 11/19/2020] [Indexed: 02/01/2023] Open
Abstract
The long pentraxin 3 (PTX3) plays a critical role in inflammation, tissue repair, and wound healing. Here, we show that PTX3 regulates disease pathogenesis in cutaneous leishmaniasis (CL). PTX3 expression increases in skin lesions in patients and mice during CL, with higher expression correlating with severe disease. PTX3-deficient (PTX3-/-) mice are highly resistant to L. major and L. braziliensis infections. This enhanced resistance is associated with increases in Th17 and IL-17A responses. The neutralization of IL-17A abolishes this enhanced resistance, while rPTX3 treatment results in decrease in Th17 and IL-17A responses and increases susceptibility. PTX3-/- CD4+ T cells display increased differentiation to Th17 and expression of Th17-specific transcription factors. The addition of rPTX3 suppresses the expression of Th17 transcription factors, Th17 differentiation, and IL-17A production by CD4+ T cells from PTX3-/- mice. Collectively, our results show that PTX3 contributes to the pathogenesis of CL by negatively regulating Th17 and IL-17A responses.
Collapse
Affiliation(s)
- Gaurav Gupta
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; NIIT University, Rajasthan, India
| | - Zhirong Mou
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Ping Jia
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Rohit Sharma
- Instituto Gonçalo Muniz (IGM), FIOCRUZ, Salvador, Brazil
| | - Romaniya Zayats
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | | | - Lianyu Shan
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Aldina Barral
- Instituto Gonçalo Muniz (IGM), FIOCRUZ, Salvador, Brazil
| | | | - Thomas T Murooka
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Abdel Soussi-Gounni
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | | | - Jude E Uzonna
- Department of Immunology, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
22
|
Bekeredjian-Ding I. Challenges for Clinical Development of Vaccines for Prevention of Hospital-Acquired Bacterial Infections. Front Immunol 2020; 11:1755. [PMID: 32849627 PMCID: PMC7419648 DOI: 10.3389/fimmu.2020.01755] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
Increasing antibiotic resistance in bacteria causing endogenous infections has entailed a need for innovative approaches to therapy and prophylaxis of these infections and raised a new interest in vaccines for prevention of colonization and infection by typically antibiotic resistant pathogens. Nevertheless, there has been a long history of failures in late stage clinical development of this type of vaccines, which remains not fully understood. This article provides an overview on present and past vaccine developments targeting nosocomial bacterial pathogens; it further highlights the specific challenges associated with demonstrating clinical efficacy of these vaccines and the facts to be considered in future study designs. Notably, these vaccines are mainly applied to subjects with preexistent immunity to the target pathogen, transient or chronic immunosuppression and ill-defined microbiome status. Unpredictable attack rates and changing epidemiology as well as highly variable genetic and immunological strain characteristics complicate the development. In views of the clinical need, re-thinking of the study designs and expectations seems warranted: first of all, vaccine development needs to be footed on a clear rationale for choosing the immunological mechanism of action and the optimal time point for vaccination, e.g., (1) prevention (or reduction) of colonization vs. prevention of infection and (2) boosting of a preexistent immune response vs. altering the quality of the immune response. Furthermore, there are different, probably redundant, immunological and microbiological defense mechanisms that provide protection from infection. Their interplay is not well-understood but as a consequence their effect might superimpose vaccine-mediated resolution of infection and lead to failure to demonstrate efficacy. This implies that improved characterization of patient subpopulations within the trial population should be obtained by pro- and retrospective analyses of trial data on subject level. Statistical and systems biology approaches could help to define immune and microbiological biomarkers that discern populations that benefit from vaccination from those where vaccines might not be effective.
Collapse
Affiliation(s)
- Isabelle Bekeredjian-Ding
- Division of Microbiology, Langen, Germany.,Institute for Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
23
|
Zhao J, Chen X, Herjan T, Li X. The role of interleukin-17 in tumor development and progression. J Exp Med 2020; 217:jem.20190297. [PMID: 31727782 PMCID: PMC7037244 DOI: 10.1084/jem.20190297] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/21/2019] [Accepted: 10/08/2019] [Indexed: 12/22/2022] Open
Abstract
IL-17, a potent proinflammatory cytokine, has been shown to intimately contribute to the formation, growth, and metastasis of a wide range of malignancies. Recent studies implicate IL-17 as a link among inflammation, wound healing, and cancer. While IL-17-mediated production of inflammatory mediators mobilizes immune-suppressive and angiogenic myeloid cells, emerging studies reveal that IL-17 can directly act on tissue stem cells to promote tissue repair and tumorigenesis. Here, we review the pleotropic impacts of IL-17 on cancer biology, focusing how IL-17-mediated inflammatory response and mitogenic signaling are exploited to equip its cancer-promoting function and discussing the implications in therapies.
Collapse
Affiliation(s)
- Junjie Zhao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xing Chen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Tomasz Herjan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
24
|
Pan X, Fan Z, Chen L, Liu C, Bai F, Wei Y, Tian Z, Dong Y, Shi J, Chen H, Jin Y, Cheng Z, Jin S, Lin J, Wu W. PvrA is a novel regulator that contributes to Pseudomonas aeruginosa pathogenesis by controlling bacterial utilization of long chain fatty acids. Nucleic Acids Res 2020; 48:5967-5985. [PMID: 32406921 PMCID: PMC7293031 DOI: 10.1093/nar/gkaa377] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 12/19/2022] Open
Abstract
During infection of a host, Pseudomonas aeruginosa orchestrates global gene expression to adapt to the host environment and counter the immune attacks. P. aeruginosa harbours hundreds of regulatory genes that play essential roles in controlling gene expression. However, their contributions to the bacterial pathogenesis remain largely unknown. In this study, we analysed the transcriptomic profile of P. aeruginosa cells isolated from lungs of infected mice and examined the roles of upregulated regulatory genes in bacterial virulence. Mutation of a novel regulatory gene pvrA (PA2957) attenuated the bacterial virulence in an acute pneumonia model. Chromatin immunoprecipitation (ChIP)-Seq and genetic analyses revealed that PvrA directly regulates genes involved in phosphatidylcholine utilization and fatty acid catabolism. Mutation of the pvrA resulted in defective bacterial growth when phosphatidylcholine or palmitic acid was used as the sole carbon source. We further demonstrated that palmitoyl coenzyme A is a ligand for the PvrA, enhancing the binding affinity of PvrA to its target promoters. An arginine residue at position 136 was found to be essential for PvrA to bind palmitoyl coenzyme A. Overall, our results revealed a novel regulatory pathway that controls genes involved in phosphatidylcholine and fatty acid utilization and contributes to the bacterial virulence.
Collapse
Affiliation(s)
- Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zheng Fan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Lei Chen
- Department of Plant Biology and Ecology, College of Life Science Nankai University, Tianjin 300071 China
| | - Chang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Fang Bai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yu Wei
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Zhenyang Tian
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuanyuan Dong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Jing Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Hao Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Shouguang Jin
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jianping Lin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
25
|
Ahmadbeigi Y, Chirani AS, Soleimani N, Mahdavi M, Goudarzi M. Immunopotentiation of the engineered low-molecular-weight pilin targeting Pseudomonas aeruginosa: A combination of immunoinformatics investigation and active immunization. Mol Immunol 2020; 124:70-82. [PMID: 32540517 DOI: 10.1016/j.molimm.2020.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 04/22/2020] [Accepted: 05/11/2020] [Indexed: 01/08/2023]
Abstract
Several vaccine candidates have been introduced for immunization against Pseudomonas aeruginosa strains. Despite extensive efforts in recent decades, there is no accurate immunogenic candidate against this pathogen in the market yet. Due to the rapid increase in several drug-resistant strains, P. aeruginosa has caused various health concerns worldwide. It encodes many specific virulence features, which can be used as an appropriate vaccine candidate. The primary stage of the pathogenesis of P. aeruginosa is the expression of many dynamic adhesive molecules, such as type IV pili (T4P), which acts as a principal colonization factor. It has been confirmed that three different subtypes of T4P, including type IVa (T4aP), type IVb (T4bP) and tight adherence (Tad) pili are expressed by P. aeruginosa. The IVa fimbriae type is almost the main cause of challenges to design an effective pili based-immunotherapy method. Nevertheless, in terms of heterogeneity, variability and hidden conserved binding site of T4aP, this attitude has been remained controversial and there is no permitted human study based on IVa pilin commercially. The engineered synthetic peptide-based vaccines are highly talented to mimic the target. In this research, for the first time, some dominant immunogenic features of the Flp protein, such as both B- and T-cell-associated epitopes, presence of IgE-associated epitopes, solvent-accessible surface area were evaluated by analytical immunoinformatics methods. In addition, we designed the engineered Flp pilin as an effective immunogenic substance against several clinically important P. aeruginosa strains. Moreover, by practical active immunization approaches, the humoral and cellular immune response against the extremely conserved region of the engineered synthetic Flp (EFlp) formulated in Montanide ISA 266 compared to the control group. The results of active immunization against EFlp significantly signified that EFlp-Montanide ISA 266 (EFLP-M) strongly could induce both humoral and cellular immune responses. We concluded that Flp pilin has therapeutic potential against numerous clinically significant P. aeruginosa strains and can be served as a novel immunogen for further investigations for development of effective immunotherapy methods against P. aeruginosa as a dexterous pathogen.
Collapse
Affiliation(s)
- Yasaman Ahmadbeigi
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Alireza Salimi Chirani
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Soleimani
- Department of Microbiology and Microbial Biotechnology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mehdi Mahdavi
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Immunotherapy Group, The Institute of Pharmaceutical Science (TIPS), Tehran University of Medical Science, Tehran, Iran; Departments of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Hlavová K, Štěpánová H, Šťastný K, Levá L, Hodkovicová N, Vícenová M, Matiašovic J, Faldyna M. Minimal Concentrations of Deoxynivalenol Reduce Cytokine Production in Individual Lymphocyte Populations in Pigs. Toxins (Basel) 2020; 12:toxins12030190. [PMID: 32197345 PMCID: PMC7150743 DOI: 10.3390/toxins12030190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 11/16/2022] Open
Abstract
Deoxynivalenol (DON) is a mycotoxin frequently found in cereals, and pigs are one of the most sensitive farm species to DON. The aim of this study was to determine the effects of DON in very low doses on peripheral blood mononuclear cells (PBMC) and on particular lymphocyte subpopulations. The cells were exposed to 1, 10 and 100 ng/mL of DON and lymphocyte viability, proliferation, and cytokine (Interleukin (IL)-1β, IL-2, IL-8, IL-17, Interferon (IFN) γ and tumor necrosis factor (TNF) α production were studied. Cells exposed to DON for 5 days in concentrations of 1 and 10 ng/mL showed higher viability compared to control cells. After 18 h of DON (100 ng/mL) exposure, a significantly lower proliferation after mitogen stimulation was observed. In contrast, an increase of spontaneous proliferation induced by DON (100 ng/mL) was detected. After DON exposure, the expression of cytokine genes decreased, with the exception of IL-1β and IL-8, which increased after 18 h exposure to 100 ng/mL of DON. Among lymphocyte subpopulations, helper T-cells and γδ T-cells exhibiting lower production of IL-17, IFNγ and TNFα were most affected by DON exposure (10 ng/mL). These findings show that subclinical doses of DON lead to changes in immune response.
Collapse
|
27
|
Cabral MP, Correia A, Vilanova M, Gärtner F, Moscoso M, García P, Vallejo JA, Pérez A, Francisco-Tomé M, Fuentes-Valverde V, Bou G. A live auxotrophic vaccine confers mucosal immunity and protection against lethal pneumonia caused by Pseudomonas aeruginosa. PLoS Pathog 2020; 16:e1008311. [PMID: 32040500 PMCID: PMC7034913 DOI: 10.1371/journal.ppat.1008311] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 02/21/2020] [Accepted: 01/06/2020] [Indexed: 12/28/2022] Open
Abstract
Pseudomonas aeruginosa is one of the leading causes of nosocomial pneumonia and its associated mortality. Moreover, extensively drug-resistant high-risk clones are globally widespread, presenting a major challenge to the healthcare systems. Despite this, no vaccine is available against this high-concerning pathogen. Here we tested immunogenicity and protective efficacy of an experimental live vaccine against P. aeruginosa pneumonia, consisting of an auxotrophic strain which lacks the key enzyme involved in D-glutamate biosynthesis, a structural component of the bacterial cell wall. As the amounts of free D-glutamate in vivo are trace substances in most cases, blockage of the cell wall synthesis occurs, compromising the growth of this strain, but not its immunogenic properties. Indeed, when delivered intranasally, this vaccine stimulated production of systemic and mucosal antibodies, induced effector memory, central memory and IL-17A-producing CD4+ T cells, and recruited neutrophils and mononuclear phagocytes into the airway mucosa. A significant improvement in mice survival after lung infection caused by ExoU-producing PAO1 and PA14 strains was observed. Nearly one third of the mice infected with the XDR high-risk clone ST235 were also protected. These findings highlight the potential of this vaccine for the control of acute pneumonia caused by this bacterial pathogen. Pseudomonas aeruginosa is an opportunistic bacterium and one of the most common causes of healthcare-associated diseases, including acute pneumonia, causing high mortality within immunocompromised hosts. Most of these infections are strikingly difficult to treat using conventional antibiotic therapies, since this microorganism displays high intrinsic resistance to a wide range of antibiotics. Moreover, to date, no vaccine is available for prevention. Here we used a mutated bacterial strain, which is unable to replicate in vivo and to cause disease, as a live vaccine against acute pneumonia caused by this pathogen. When applied intranasally, this vaccine induced immunity both at local and distant body sites, activating immune cells which were recruited into the airway mucosa. This evoked immune response reduced the number of non-surviving mice after infection with two cytotoxic P. aeruginosa strains causing acute lung infection. Some protection was also observed against an internationally disseminated cytotoxic strain. These data indicate that this is a promising vaccine candidate against P. aeruginosa-pneumonia.
Collapse
Affiliation(s)
- Maria P. Cabral
- Department of Microbiology, University Hospital A Coruña (CHUAC)–Biomedical Research Institute A Coruña (INIBIC), A Coruña, Spain
| | - Alexandra Correia
- i3S –Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Manuel Vilanova
- i3S –Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC–Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Fátima Gärtner
- i3S –Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Miriam Moscoso
- Department of Microbiology, University Hospital A Coruña (CHUAC)–Biomedical Research Institute A Coruña (INIBIC), A Coruña, Spain
| | - Patricia García
- Department of Microbiology, University Hospital A Coruña (CHUAC)–Biomedical Research Institute A Coruña (INIBIC), A Coruña, Spain
| | - Juan A. Vallejo
- Department of Microbiology, University Hospital A Coruña (CHUAC)–Biomedical Research Institute A Coruña (INIBIC), A Coruña, Spain
| | - Astrid Pérez
- Department of Microbiology, University Hospital A Coruña (CHUAC)–Biomedical Research Institute A Coruña (INIBIC), A Coruña, Spain
| | - Mónica Francisco-Tomé
- Department of Microbiology, Galicia Sur Health Research Institute (IISGS), Vigo, Spain
| | - Víctor Fuentes-Valverde
- Department of Microbiology, University Hospital A Coruña (CHUAC)–Biomedical Research Institute A Coruña (INIBIC), A Coruña, Spain
| | - Germán Bou
- Department of Microbiology, University Hospital A Coruña (CHUAC)–Biomedical Research Institute A Coruña (INIBIC), A Coruña, Spain
- * E-mail:
| |
Collapse
|
28
|
Zhao J, Chen X, Herjan T, Li X. The role of interleukin-17 in tumor development and progression. J Exp Med 2020; 217:e20190297. [PMID: 31727782 DOI: 10.1084/jem_20190297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 08/21/2019] [Accepted: 10/08/2019] [Indexed: 01/03/2025] Open
Abstract
IL-17, a potent proinflammatory cytokine, has been shown to intimately contribute to the formation, growth, and metastasis of a wide range of malignancies. Recent studies implicate IL-17 as a link among inflammation, wound healing, and cancer. While IL-17-mediated production of inflammatory mediators mobilizes immune-suppressive and angiogenic myeloid cells, emerging studies reveal that IL-17 can directly act on tissue stem cells to promote tissue repair and tumorigenesis. Here, we review the pleotropic impacts of IL-17 on cancer biology, focusing how IL-17-mediated inflammatory response and mitogenic signaling are exploited to equip its cancer-promoting function and discussing the implications in therapies.
Collapse
Affiliation(s)
- Junjie Zhao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xing Chen
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Tomasz Herjan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
29
|
Sen-Kilic E, Blackwood CB, Boehm DT, Witt WT, Malkowski AC, Bevere JR, Wong TY, Hall JM, Bradford SD, Varney ME, Damron FH, Barbier M. Intranasal Peptide-Based FpvA-KLH Conjugate Vaccine Protects Mice From Pseudomonas aeruginosa Acute Murine Pneumonia. Front Immunol 2019; 10:2497. [PMID: 31708925 PMCID: PMC6819369 DOI: 10.3389/fimmu.2019.02497] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing acute and chronic respiratory infections associated with morbidity and mortality, especially in patients with cystic fibrosis. Vaccination against P. aeruginosa before colonization may be a solution against these infections and improve the quality of life of at-risk patients. To develop a vaccine against P. aeruginosa, we formulated a novel peptide-based P. aeruginosa subunit vaccine based on the extracellular regions of one of its major siderophore receptors, FpvA. We evaluated the effectiveness and immunogenicity of the FpvA peptides conjugated to keyhole limpet hemocyanin (KLH) with the adjuvant curdlan in a murine vaccination and challenge model. Immunization with the FpvA-KLH vaccine decreased the bacterial burden and lung edema after P. aeruginosa challenge. Vaccination with FpvA-KLH lead to antigen-specific IgG and IgM antibodies in sera, and IgA antibodies in lung supernatant. FpvA-KLH immunized mice had an increase in recruitment of CD11b+ dendritic cells as well as resident memory CD4+ T cells in the lungs compared to non-vaccinated challenged mice. Splenocytes isolated from vaccinated animals showed that the FpvA-KLH vaccine with the adjuvant curdlan induces antigen-specific IL-17 production and leads to a Th17 type of immune response. These results indicate that the intranasal FpvA-KLH conjugate vaccine can elicit both mucosal and systemic immune responses. These observations suggest that the intranasal peptide-based FpvA-KLH conjugate vaccine with curdlan is a potential vaccine candidate against P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Catherine B Blackwood
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Dylan T Boehm
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - William T Witt
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Aaron C Malkowski
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Justin R Bevere
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Ting Y Wong
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Jesse M Hall
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Shelby D Bradford
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Melinda E Varney
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Fredrick Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States.,Vaccine Development Center at West Virginia University Health Sciences Center, Morgantown, WV, United States
| |
Collapse
|
30
|
Ranjbar M, Behrouz B, Norouzi F, Mousavi Gargari SL. Anti-PcrV IgY antibodies protect against Pseudomonas aeruginosa infection in both acute pneumonia and burn wound models. Mol Immunol 2019; 116:98-105. [PMID: 31634816 DOI: 10.1016/j.molimm.2019.10.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/11/2022]
Abstract
Pseudomonas aeruginosa is a common nosocomial pathogen in burn patients, and rapidly acquires antibiotic resistance; thus, developing an effective therapeutic approach is the most promising strategy for combating infection. Type III secretion system (T3SS) translocates bacterial toxins into the cytosol of the targeted eukaryotic cells, which plays important roles in the virulence of P. aeruginosa infections in both acute pneumonia and burn wound models. The PcrV protein, a T3SS translocating protein, is required for T3SS function and is a well-validated target in animal models of immunoprophylactic strategies targeting P. aeruginosa. In the present study, we evaluated the protective efficacy of chicken egg yolk antibodies (IgY) raised against recombinant PcrV (r-PcrV) in both acute pneumonia and burn wound models. R-PcrV protein was generated by expressing the pcrV gene (cloned in pET-28a vector) in E. coli BL-21. Anti-PcrV IgY was obtained by immunization of hen. Anti-PcrV IgY induced greater protection in P. aeruginosamurine acute pneumonia and burn wound models than control IgY (C-IgY) and PBS groups. Anti-PcrV IgY improved opsonophagocytic killing and inhibition of bacterial invasion of host cells. Taken together, our data provide evidence that anti-PcrV IgY can be a promising therapeutic candidate for combating P. aeruginosa infections.
Collapse
Affiliation(s)
- Mahya Ranjbar
- Department of Microbiology, Shahed University, Faculty of Medical Sciences, Tehran, Iran; Department of Biology, Faculty of Basic Science, Shahed University, Tehran, Iran
| | - Bahador Behrouz
- Department of Biology, Faculty of Basic Science, Shahed University, Tehran, Iran
| | - Fatemeh Norouzi
- Department of Biology, Faculty of Basic Science, Shahed University, Tehran, Iran
| | | |
Collapse
|
31
|
Baker SM, McLachlan JB, Morici LA. Immunological considerations in the development of Pseudomonas aeruginosa vaccines. Hum Vaccin Immunother 2019; 16:412-418. [PMID: 31368828 DOI: 10.1080/21645515.2019.1650999] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogen capable of causing a wide range of potentially life-threatening infections. With multidrug-resistant P. aeruginosa infections on the rise, the need for a rationally-designed vaccine against this pathogen is critical. A number of vaccine platforms have shown promising results in pre-clinical studies, but no vaccine has successfully advanced to licensure. Growing evidence suggests that an effective P. aeruginosa vaccine may require Th17-type CD4+ T cells to prevent infection. In this review, we summarize recent pre-clinical studies of P. aeruginosa vaccines, specifically focusing on those that induce Th17-type cellular immunity. We also highlight the importance of adjuvant selection and immunization route in vaccine design in order to target vaccine-induced immunity to infected tissues. Advances in cellular immunology and adjuvant biology may ultimately influence better P. aeruginosa vaccine platforms that can protect targeted human populations.
Collapse
Affiliation(s)
- Sarah M Baker
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - James B McLachlan
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lisa A Morici
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
32
|
Wei X, Ran D, Campeau A, Xiao C, Zhou J, Dehaini D, Jiang Y, Kroll AV, Zhang Q, Gao W, Gonzalez DJ, Fang RH, Zhang L. Multiantigenic Nanotoxoids for Antivirulence Vaccination against Antibiotic-Resistant Gram-Negative Bacteria. NANO LETTERS 2019. [PMID: 31184899 DOI: 10.1021/acs.nanolett.9b0184410.1021/acs.nanolett.9b01844.s001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Infections caused by multidrug-resistant Gram-negative bacteria have emerged as a major threat to public health worldwide. The high mortality and prevalence, along with the slow pace of new antibiotic discovery, highlight the necessity for new disease management paradigms. Here, we report on the development of a multiantigenic nanotoxoid vaccine based on macrophage membrane-coated nanoparticles for eliciting potent immunity against pathogenic Pseudomonas aeruginosa. The design of this biomimetic nanovaccine leverages the specific role of macrophages in clearing pathogens and their natural affinity for various virulence factors secreted by the bacteria. It is demonstrated that the macrophage nanotoxoid is able to display a wide range of P. aeruginosa antigens, and the safety of the formulation is confirmed both in vitro and in vivo. When used to vaccinate mice via different administration routes, the nanotoxoid is capable of eliciting strong humoral immune responses that translate into enhanced protection against live bacterial infection in a pneumonia model. Overall, the work presented here provides new insights into the design of safe, multiantigenic antivirulence vaccines using biomimetic nanotechnology and the application of these nanovaccines toward the prevention of difficult-to-treat Gram-negative infections.
Collapse
Affiliation(s)
- Xiaoli Wei
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Danni Ran
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Anaamika Campeau
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences , University of California San Diego , La Jolla , California 92093 , United States
| | - Crystal Xiao
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Jiarong Zhou
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Diana Dehaini
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Yao Jiang
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Ashley V Kroll
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Qiangzhe Zhang
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Weiwei Gao
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - David J Gonzalez
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences , University of California San Diego , La Jolla , California 92093 , United States
| | - Ronnie H Fang
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| | - Liangfang Zhang
- Department of NanoEngineering and Chemical Engineering Program , University of California San Diego , La Jolla , California 92093 , United States
- Moores Cancer Center , University of California San Diego , La Jolla , California 92093 , United States
| |
Collapse
|
33
|
Wei X, Ran D, Campeau A, Xiao C, Zhou J, Dehaini D, Jiang Y, Kroll AV, Zhang Q, Gao W, Gonzalez DJ, Fang RH, Zhang L. Multiantigenic Nanotoxoids for Antivirulence Vaccination against Antibiotic-Resistant Gram-Negative Bacteria. NANO LETTERS 2019; 19:4760-4769. [PMID: 31184899 PMCID: PMC6711367 DOI: 10.1021/acs.nanolett.9b01844] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Infections caused by multidrug-resistant Gram-negative bacteria have emerged as a major threat to public health worldwide. The high mortality and prevalence, along with the slow pace of new antibiotic discovery, highlight the necessity for new disease management paradigms. Here, we report on the development of a multiantigenic nanotoxoid vaccine based on macrophage membrane-coated nanoparticles for eliciting potent immunity against pathogenic Pseudomonas aeruginosa. The design of this biomimetic nanovaccine leverages the specific role of macrophages in clearing pathogens and their natural affinity for various virulence factors secreted by the bacteria. It is demonstrated that the macrophage nanotoxoid is able to display a wide range of P. aeruginosa antigens, and the safety of the formulation is confirmed both in vitro and in vivo. When used to vaccinate mice via different administration routes, the nanotoxoid is capable of eliciting strong humoral immune responses that translate into enhanced protection against live bacterial infection in a pneumonia model. Overall, the work presented here provides new insights into the design of safe, multiantigenic antivirulence vaccines using biomimetic nanotechnology and the application of these nanovaccines toward the prevention of difficult-to-treat Gram-negative infections.
Collapse
Affiliation(s)
- Xiaoli Wei
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Danni Ran
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Anaamika Campeau
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093
| | - Crystal Xiao
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Jiarong Zhou
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Diana Dehaini
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Yao Jiang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Ashley V. Kroll
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Qiangzhe Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Weiwei Gao
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - David J. Gonzalez
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093
| | - Ronnie H. Fang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| | - Liangfang Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, CA 92093
- Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
34
|
Wan C, Zhang J, Zhao L, Cheng X, Gao C, Wang Y, Xu W, Zou Q, Gu J. Rational Design of a Chimeric Derivative of PcrV as a Subunit Vaccine Against Pseudomonas aeruginosa. Front Immunol 2019; 10:781. [PMID: 31068928 PMCID: PMC6491502 DOI: 10.3389/fimmu.2019.00781] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/25/2019] [Indexed: 01/20/2023] Open
Abstract
Pseudomonas aeruginosa (PA) is a major cause of nosocomial infections, which remain an unsolved problem in the clinic despite conventional antibiotic treatment. A PA vaccine could be both an effective and economical strategy to address this issue. Many studies have shown that PcrV, a structural protein of the type 3 secretion system (T3SS) from PA, is an ideal target for immune prevention and therapy. However, difficulties in the production of high-quality PcrV likely hinder its further application in the vaccine industry. Thus, we hypothesized that an optimized PcrV derivative with a rational design could be produced. In this study, the full-length PcrV was divided into four domains with the guidance of its structure, and the Nter domain (Met1-Lys127) and H12 domain (Leu251-Ile294) were found to be immunodominant. Subsequently, Nter and H12 were combined with a flexible linker to generate an artificial PcrV derivative (PcrVNH). PcrVNH was successfully produced in E. coli and behaved as a homogenous monomer. Moreover, immunization with PcrVNH elicited a multifactorial immune response and conferred broad protection in an acute PA pneumonia model and was equally effective to full-length PcrV. In addition, passive immunization with anti-PcrVNH antibodies alone also showed significant protection, at least based on inhibition of the T3SS and mediation of opsonophagocytic killing activities. These results provide an additional example for the rational design of antigens and suggest that PcrVNH is a promising vaccine candidate for the control of PA infection.
Collapse
Affiliation(s)
- Chuang Wan
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jin Zhang
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
- Department of Critical Care Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Liqun Zhao
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xin Cheng
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Chen Gao
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ying Wang
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wanting Xu
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
- Department of Critical Care Medicine, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Quanming Zou
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jiang Gu
- Department of Microbiology and Biochemical Pharmacy, National Engineering Research Center of Immunological Products, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
35
|
Fan Y, Mu Y, Lu L, Tian Y, Yuan F, Zhou B, Yu C, Wang Z, Li X, Lei S, Xu Y, Wu D, Yang L. Hydrogen peroxide-inactivated bacteria induces potent humoral and cellular immune responses and releases nucleic acids. Int Immunopharmacol 2019; 69:389-397. [DOI: 10.1016/j.intimp.2019.01.055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/24/2019] [Accepted: 01/31/2019] [Indexed: 01/07/2023]
|
36
|
Hoggarth A, Weaver A, Pu Q, Huang T, Schettler J, Chen F, Yuan X, Wu M. Mechanistic research holds promise for bacterial vaccines and phage therapies for Pseudomonas aeruginosa. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:909-924. [PMID: 30936684 PMCID: PMC6431001 DOI: 10.2147/dddt.s189847] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vaccines for Pseudomonas aeruginosa have been of longstanding interest to immunologists, bacteriologists, and clinicians, due to the widespread prevalence of hospital-acquired infection. As P. aeruginosa becomes increasingly antibiotic resistant, there is a dire need for novel treatments and preventive vaccines. Despite intense efforts, there currently remains no vaccine on the market to combat this dangerous pathogen. This article summarizes current and past vaccines under development that target various constituents of P. aeruginosa. Targeting lipopolysaccharides and O-antigens have shown some promise in preventing infection. Recombinant flagella and pili that target TLR5 have been utilized to combat P. aeruginosa by blocking its motility and adhesion. The type 3 secretion system components, such as needle-like structure PcrV or exotoxin PopB, are also potential vaccine targets. Outer membrane proteins including OprF and OprI are newer representatives of vaccine candidates. Live attenuated vaccines are a focal point in this review, and are also considered for novel vaccines. In addition, phage therapy is revived as an effective option for treating refractory infections after failure with antibiotic treatment. Many of the aforementioned vaccines act on a single target, thus lacking a broad range of protection. Recent studies have shown that mixtures of vaccines and combination approaches may significantly augment immunogenicity, thereby increasing their preventive and therapeutic potential.
Collapse
Affiliation(s)
- Austin Hoggarth
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| | - Andrew Weaver
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| | - Qinqin Pu
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| | - Ting Huang
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA, .,Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Jacob Schettler
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| | - Feng Chen
- Pulmonary and Allergy Institute, Affiliated Hospital of Southwestern Medical University, Luzhou, China
| | - Xiefang Yuan
- Pulmonary and Allergy Institute, Affiliated Hospital of Southwestern Medical University, Luzhou, China
| | - Min Wu
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA,
| |
Collapse
|
37
|
Baker SM, Pociask D, Clements JD, McLachlan JB, Morici LA. Intradermal vaccination with a Pseudomonas aeruginosa vaccine adjuvanted with a mutant bacterial ADP-ribosylating enterotoxin protects against acute pneumonia. Vaccine 2019; 37:808-816. [PMID: 30638799 DOI: 10.1016/j.vaccine.2018.12.053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 12/20/2022]
Abstract
Respiratory infections are a leading cause of morbidity and mortality globally. This is partially due to a lack of effective vaccines and a clear understanding of how vaccination route and formulation influence protective immunity in mucosal tissues such as the lung. Pseudomonas aeruginosa is an opportunistic pathogen capable of causing acute pulmonary infections and is a leading cause of hospital-acquired and ventilator-associated pneumonia. With multidrug-resistant P. aeruginosa infections on the rise, the need for a vaccine against this pathogen is critical. Growing evidence suggests that a successful P. aeruginosa vaccine may require mucosal antibody and Th1- and Th17-type CD4+ T cells to prevent pulmonary infection. Intradermal immunization with adjuvants, such as the bacterial ADP-Ribosylating Enterotoxin Adjuvant (BARE) double mutant of E. coli heat-labile toxin (dmLT), can direct protective immune responses to mucosal tissues, including the lungs. We reasoned that intradermal immunization with P. aeruginosa outer membrane proteins (OMPs) adjuvanted with dmLT could drive neutralizing antibodies and migration of CD4+ T cells to the lungs and protect against P. aeruginosa pneumonia in a murine model. Here we show that mice immunized with OMPs and dmLT had significantly more antigen-specific IgG and Th1- and Th17-type CD4+ memory T cells in the pulmonary environment compared to control groups of mice. Furthermore, OMPs and dmLT immunized mice were significantly protected against an otherwise lethal lung infection. Protection was associated with early IFN-γ and IL-17 production in the lungs of immunized mice. These results indicate that intradermal immunization with dmLT can drive protective immunity to the lung mucosa and may be a viable vaccination strategy for a multitude of respiratory pathogens.
Collapse
Affiliation(s)
- Sarah M Baker
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA
| | - Derek Pociask
- Department of Medicine, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA
| | - John D Clements
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA
| | - James B McLachlan
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA
| | - Lisa A Morici
- Department of Microbiology and Immunology, School of Medicine, Tulane University, 1430 Tulane Ave., New Orleans, LA, USA.
| |
Collapse
|
38
|
Merakou C, Schaefers MM, Priebe GP. Progress Toward the Elusive Pseudomonas aeruginosa Vaccine. Surg Infect (Larchmt) 2018; 19:757-768. [PMID: 30388058 DOI: 10.1089/sur.2018.233] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: The gram-negative bacterial pathogen Pseudomonas aeruginosa causes a wide range of infections, mostly in hospitalized and immunocompromised patients, those with burns, surgical wounds, or combat-related wounds, and in people with cystic fibrosis. The increasing antibiotic resistance of P. aeruginosa confers a pressing need for vaccines, yet there are no P. aeruginosa vaccines approved for human use, and recent promising candidates have failed in large clinical trials. Discussion: In this review, we summarize recent clinical trials and pre-clinical studies of P. aeruginosa vaccines and provide a suggested framework for the makeup of a future successful vaccine. Murine models of infection suggest that antibodies, specifically opsonophagocytic killing antibodies (OPK), antitoxin antibodies, and anti-attachment antibodies, combined with T cell immunity, specifically TH17 responses, are needed for broad and potent protection against P. aeruginosa infection. A better understanding of the human immune response to P. aeruginosa infections, and to vaccine candidates, will eventually pave the way to a successful vaccine for this wily pathogen.
Collapse
Affiliation(s)
- Christina Merakou
- 1 Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital , Boston, Massachusetts.,2 Department of Anaesthesia, Harvard Medical School , Boston, Massachusetts
| | - Matthew M Schaefers
- 1 Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital , Boston, Massachusetts.,2 Department of Anaesthesia, Harvard Medical School , Boston, Massachusetts
| | - Gregory P Priebe
- 1 Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital , Boston, Massachusetts.,2 Department of Anaesthesia, Harvard Medical School , Boston, Massachusetts.,3 Division of Infectious Diseases, Department of Pediatrics, Boston Children's Hospital , Boston, Massachusetts
| |
Collapse
|
39
|
Schaefers MM, Duan B, Mizrahi B, Lu R, Reznor G, Kohane DS, Priebe GP. PLGA-encapsulation of the Pseudomonas aeruginosa PopB vaccine antigen improves Th17 responses and confers protection against experimental acute pneumonia. Vaccine 2018; 36:6926-6932. [PMID: 30314911 DOI: 10.1016/j.vaccine.2018.10.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/20/2018] [Accepted: 10/03/2018] [Indexed: 12/26/2022]
Abstract
The Pseudomonas aeruginosa type III secretion system protein PopB and its chaperon protein PcrH, when co-administered with the adjuvant curdlan, elicit Th17 responses after intranasal immunization of mice. These PopB/PcrH-curdlan vaccines protect mice against acute lethal pneumonia in an IL-17-dependent fashion involving CD4 helper T cells secreting IL-17 (Th17 cells). In this study, we tested whether encapsulation of PopB/PcrH in poly-lactic-co-glycolic acid (PLGA) nanoparticles could elicit Th17 responses to PopB. Recombinant PopB/PcrH or PcrH alone was encapsulated into PLGA nanoparticles. Mice (FVB/N) were intranasally immunized with the PLGA-PopB/PcrH nanoparticles, PLGA-PcrH nanoparticles, PLGA alone, or PopB/PcrH alone. The protective efficacy was assessed in an acute lung infection model with a lethal dose of an ExoU-producing version of P. aeruginosa strain PAO1. Th17 responses were assayed by intracellular flow cytometry and by ELISA for IL-17 in supernatants of splenocytes co-cultured with purified PopB/PcrH. PLGA-PopB/PcrH-immunized mice showed 3-4-fold higher Th17 responses both in the lung and in the spleen compared to mice immunized with empty PLGA or PopB/PcrH alone. After challenge with P. aeruginosa, PLGA-PopB/PcrH-immunized mice showed significantly lower bacterial counts in the lungs and improved survival. In conclusion, encapsulation of PopB/PcrH in PLGA nanoparticles can elicit Th17 responses to intranasal vaccination and protect mice against acute lethal P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Matthew M Schaefers
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115 USA; Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA.
| | - Biyan Duan
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA
| | - Boaz Mizrahi
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Roger Lu
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115 USA; Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA
| | - Gally Reznor
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Daniel S Kohane
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115 USA; Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Gregory P Priebe
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115 USA; Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA
| |
Collapse
|
40
|
Wang S, Gao J, Li M, Wang L, Wang Z. A facile approach for development of a vaccine made of bacterial double-layered membrane vesicles (DMVs). Biomaterials 2018; 187:28-38. [PMID: 30292939 DOI: 10.1016/j.biomaterials.2018.09.042] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/15/2018] [Accepted: 09/28/2018] [Indexed: 12/13/2022]
Abstract
Bacterial infections cause acute and chronic diseases. Antimicrobial resistance and aging-related immune weakness remain challenging in therapy of infectious diseases. Vaccines are however an alternative to prevent bacterial infections. Here we report a facile method to rapidly generate bacterium-membrane-formed nanovesicles as a vaccine using nitrogen cavitation. The vaccine is comprised of double-layered membrane vesicles (DMVs) characterized by cryo-TEM, biochemistry and proteomics, showing DMVs possess the integrity of bacterial membrane and contain a wide range of membrane proteins required for vaccination. In the mouse sepsis model induced by Pseudomonas aeruginosa, we found that DMVs can improve mouse survival after mice were immunized with DMVs. The increased adaptive immunity and unique biodistribution of DMVs were responsible for enhanced protection of bacterial infection. Our studies demonstrate that this simple and innovative approach using nitrogen cavitation would be a promising technology for vaccine developments.
Collapse
Affiliation(s)
- Sihan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Jin Gao
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA
| | - Mo Li
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Liguo Wang
- Department of Biological Structure, School of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Zhenjia Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
41
|
Immunization with Pseudomonas aeruginosa outer membrane vesicles stimulates protective immunity in mice. Vaccine 2018; 36:1047-1054. [PMID: 29406241 DOI: 10.1016/j.vaccine.2018.01.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/06/2017] [Accepted: 01/11/2018] [Indexed: 11/24/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen responsible for a wide range of severe nosocomial and community acquired infections, these infections are major health problems for cystic fibrosis patients and immune-compromised individuals. The emergence of multidrug-resistant isolates highlights the need to develop alternative strategies for treatment of P. aeruginosa infections. Outer membrane vesicles (OMVs) are spherical nanometer-sized proteolipids that are secreted from numerous of pathogenic Gram-negative bacteria, and a number of studies have confirmed the protective efficacy for use of OMVs as candidate vaccines. In this study, OMVs from P. aeruginosa (PA_OMVs) were isolated, formulated with aluminum phosphate adjuvant and used as a vaccine in a mouse model of acute lung infection. The results confirmed that active immunization with PA_OMVs was able to reduce bacterial colonization, cytokine secretion and tissue damage in the lung tissue, thus protecting mice from lethal challenge of P. aeruginosa. Cytokines assay validated that immunization with PA_OMVs was efficient to induce a mixed cellular immune response in mice. Further, high level of specific antibodies was detected in mice immunized with PA_OMVs, and results from opsonophagocytic killing assay and passive immunization suggested that humoral immune response may be critical for PA_OMVs mediated protection. These findings demonstrated that PA_OMVs may be served as a novel candidate vaccine for the prevention of P. aeruginosa infection.
Collapse
|
42
|
Bakht Azad S, Nikokar I, Faezi S, Rasooly S, Mahdavi M. Evaluation of the immune responses following co-administration of PilQ and type b-flagellin from Pseudomonas aeruginosa in the burn mouse model. Microb Pathog 2018; 123:426-432. [PMID: 30075242 DOI: 10.1016/j.micpath.2018.07.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 07/16/2018] [Accepted: 07/30/2018] [Indexed: 12/23/2022]
Abstract
Considering the increased antibiotic resistance of Pseudomonas aeruginosa, the evaluation of immune response against the antigens of this bacterium seems necessary. In this study, the protective efficacy and immunological properties of P. aeruginosa recombinant PilQ (r-PilQ) and type b-flagellin (FLB) proteins was evaluated in the burn mouse model of infection. The inbred BALB/c mice were immunized with r-PilQ and FLB antigens. To investigate the type of induced immune response, sera were analyzed by ELISA for total IgG, IgG1, and IgG2a isotypes. After the final immunization, the IL-4, IFN-γ, and IL-17 cytokines level were examined in the spleen of non-challenged mice. Fifty days after lethal challenge, the survival rate and bacterial burden in the skin and other internal organs of experimental mice were assessed. The in vivo administration of r-PilQ, FLB and combined antigen resulted in a significant increase in the survival of mice (66%, 75%, and 83%, respectively) infected by the PAO1 strain of P. aeruginosa in the burn model of infection. Immunization of mice with r-PilQ and FLB mixture induced high titers of IL-4 and IL-17 cytokines compared to control groups (P < 0.05). The high titer of antisera raised against combined antigen was able to inhibit the systemic spread of the PAO1 strain from the site of infection to the internal organs. We concluded that the parallel role of IL-4 and IL-17 is necessary for elimination of the bacteria and promotion of survival in the immunized burn mice.
Collapse
Affiliation(s)
- Sima Bakht Azad
- Medical Biotechnology Research Center, Laboratory of Microbiology and Immunology of Infectious Diseases, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Iraj Nikokar
- Medical Biotechnology Research Center, Laboratory of Microbiology and Immunology of Infectious Diseases, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran; Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran.
| | - Sobhan Faezi
- Medical Biotechnology Research Center, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Soheila Rasooly
- Medical Biotechnology Research Center, Laboratory of Microbiology and Immunology of Infectious Diseases, Paramedicine Faculty, Guilan University of Medical Sciences, Rasht, Iran
| | - Mehdi Mahdavi
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
43
|
Liu C, Pan X, Xia B, Chen F, Jin Y, Bai F, Priebe G, Cheng Z, Jin S, Wu W. Construction of a Protective Vaccine Against Lipopolysaccharide-Heterologous Pseudomonas aeruginosa Strains Based on Expression Profiling of Outer Membrane Proteins During Infection. Front Immunol 2018; 9:1737. [PMID: 30093906 PMCID: PMC6070602 DOI: 10.3389/fimmu.2018.01737] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 07/13/2018] [Indexed: 12/20/2022] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous opportunistic pathogen, which causes infectious disease in patients with cystic fibrosis and compromised immunity. P. aeruginosa is difficult to eradicate because of its intrinsic resistance to most traditional antibiotics as well as acquired resistance mechanisms after decades of antibiotic usage. A full understanding of the P. aeruginosa pathogenesis mechanisms is necessary for the development of novel prevention and treatment strategies. To identify novel vaccine candidates, here we comprehensively examined the expression levels of all the known outer membrane proteins in two P. aeruginosa strains in a murine acute pneumonia model. OprH was one of the most highly expressed proteins during infection. In addition, OprH is known to be highly immunogenic and accessible by host proteins. Thus, it was chosen as a vaccine candidate. To further identify vaccine candidates, 34 genes highly expressed during infection were evaluated for their contributions in virulence by testing individual transposon insertion mutants. Among them, fpvA, hasR, and foxA were found essential for bacterial virulence and therefore included in vaccine construction. Immunization with a mixture of FpvA, HasR, and FoxA rendered no protection, however, while immunization by OprH refolded in liposomes elicited specific opsonic antibodies and conferred protection against two lipopolysaccharide-heterologous P. aeruginosa strains (PA14 and PA103). Overall, by studying the expression profile of the P. aeruginosa outer membrane proteins during infection, we identified OprH as a potential vaccine candidate for the prevention of lung infection by P. aeruginosa.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaolei Pan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Bin Xia
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Fei Chen
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yongxin Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Fang Bai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Gregory Priebe
- Division of Critical Care Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Boston, MA, United States.,Department of Anaesthesia, Harvard Medical School, Boston, MA, United States
| | - Zhihui Cheng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Shouguang Jin
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China.,Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Weihui Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
44
|
Chauhan N, Khatri V, Banerjee P, Kalyanasundaram R. Evaluating the Vaccine Potential of a Tetravalent Fusion Protein ( rBmHAXT) Vaccine Antigen Against Lymphatic Filariasis in a Mouse Model. Front Immunol 2018; 9:1520. [PMID: 30013570 PMCID: PMC6036175 DOI: 10.3389/fimmu.2018.01520] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/19/2018] [Indexed: 12/22/2022] Open
Abstract
Lymphatic filariasis (LF) is a tropical parasitic infection of human transmitted by mosquitoes. Chronic infection results in severe physical disability in the infected patients. Although several potential vaccine antigens were identified by several groups, there are no licensed prophylactic vaccine to date against this infection in the human. Previous attempts from our laboratory to develop a trivalent prophylactic vaccine against LF showed that >90% protection could be achieved in rodent models. However, this trivalent vaccine gave only 35% protection in non-human primates. The major focus of this study was to develop a tetravalent prophylactic vaccine (rBmHAXT) and test the vaccine potential in a mouse model. We evaluated three different adjuvant formulations; alum, glucopyranosyl lipid adjuvant in stable emulsion (GLA/SE) alum (AL019), and mannosylated chitosan (MCA) to determine the optimum adjuvant formulation for rBmHAXT. Results presented in this study show that rBmHAXT + AL019 gave the highest rate of protection (>88%) against challenge infection, compared to rBmHAXT + AL007 (79%), rBmHAXT + MCA (79%) and controls. Analysis of the immune correlates of protection showed that all three adjuvants elicited high titer of antigen-specific IgG1, IgG2a, and IgG2b antibodies. High number of IFN-γ-producing antigen-specific memory cells were generated in the vaccinated animals irrespective of the adjuvants used. Similarly, spleen cells from rBmHAXT-vaccinated animals secreted IL-4, IL-10, and IFN-γ in response to rBmHAXT suggesting the generation of a balanced Th1/Th2 response. There was also an increase in IL-17-secreting cells in rBmHAXT-vaccinated animals. These findings thus suggest that rBmHAXT + AL019 is a better prophylactic formulation for LF.
Collapse
Affiliation(s)
- Nikhil Chauhan
- Department of Biomedical Sciences, University of Illinois, Rockford, IL, United States
| | - Vishal Khatri
- Department of Biomedical Sciences, University of Illinois, Rockford, IL, United States
| | - Priyankana Banerjee
- Department of Biomedical Sciences, University of Illinois, Rockford, IL, United States
| | | |
Collapse
|
45
|
Meynet E, Laurin D, Lenormand JL, Camara B, Toussaint B, Le Gouëllec A. Killed but metabolically active Pseudomonas aeruginosa-based vaccine induces protective humoral- and cell-mediated immunity against Pseudomonas aeruginosa pulmonary infections. Vaccine 2018; 36:1893-1900. [PMID: 29506924 DOI: 10.1016/j.vaccine.2018.02.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 11/20/2017] [Accepted: 02/07/2018] [Indexed: 12/17/2022]
Abstract
Pseudomonas aeruginosa (Pa) is a significant cause of morbidity and mortality, especially in cystic fibrosis patients. Its eradication is difficult due to a wide phenotypic adaptability and an increase of its resistance to antibiotics. After the failure of several recombinant vaccines which mainly triggered humoral response, live-attenuated vaccines received attention thanks to their ability to elicit a broad immunity with both humoral- and cell-mediated responses, essential to fight this pathogen. In this study, we developed an innovative and safer live-attenuated Pa vaccine based on a Killed But Metabolically Active (KBMA) attenuation method. KBMA Pa has been further rationally designed to overexpress beneficial effectors like the type 3 secretion system apparatus. We demonstrated that KBMA Pa elicits a high and broad humoral response in mice against several antigens of particular interest such as OprF and PcrV proteins. Moreover, we assessed cytokines in the serum of immunized mice and showed that KBMA Pa elicits Th1, Th2 and especially Th17 pathways of cell-mediated immune responses. Th17 pathway involvement was also confirmed after specific stimulation of helper T cells in immunized mice. Finally, we showed that this vaccine is safe and has a protective effect in a murine acute pulmonary infectious challenge. In conclusion, KBMA Pa is a new platform with high potential for the development of a vaccine against Pa.
Collapse
Affiliation(s)
- Elodie Meynet
- Univ Grenoble Alpes, CNRS, CHU Grenoble, Grenoble INP, TIMC-IMAG UMR 5525, 38000 Grenoble, France
| | - David Laurin
- Univ Grenoble Alpes, CNRS, CHU Grenoble, Grenoble INP, TIMC-IMAG UMR 5525, 38000 Grenoble, France; Etablissement Français du Sang, BP35, 38701 La Tronche, France
| | - Jean Luc Lenormand
- Univ Grenoble Alpes, CNRS, CHU Grenoble, Grenoble INP, TIMC-IMAG UMR 5525, 38000 Grenoble, France
| | - Boubou Camara
- Univ Grenoble Alpes, CNRS, CHU Grenoble, Grenoble INP, TIMC-IMAG UMR 5525, 38000 Grenoble, France
| | - Bertrand Toussaint
- Univ Grenoble Alpes, CNRS, CHU Grenoble, Grenoble INP, TIMC-IMAG UMR 5525, 38000 Grenoble, France
| | - Audrey Le Gouëllec
- Univ Grenoble Alpes, CNRS, CHU Grenoble, Grenoble INP, TIMC-IMAG UMR 5525, 38000 Grenoble, France.
| |
Collapse
|
46
|
|
47
|
Transcervical Inoculation with Chlamydia trachomatis Induces Infertility in HLA-DR4 Transgenic and Wild-Type Mice. Infect Immun 2017; 86:IAI.00722-17. [PMID: 29038126 DOI: 10.1128/iai.00722-17] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 10/10/2017] [Indexed: 12/27/2022] Open
Abstract
Chlamydia trachomatis is the leading cause of infection-induced infertility in women. Attempts to control this epidemic with screening programs and antibiotic therapy have failed. Currently, a vaccine to prevent C. trachomatis infections is not available. In order to develop an animal model for evaluating vaccine antigens that can be applied to humans, we used C. trachomatis serovar D (strain UW-3/Cx) to induce infertility in mice whose major histocompatibility complex class II antigen was replaced with the human leukocyte antigen DR4 (HLA-DR4). Transcervical inoculation of medroxyprogesterone-treated HLA-DR4 transgenic mice with 5 × 105C. trachomatis D inclusion forming units (IFU) induced a significant reduction in fertility, with a mean number of embryos/mouse of 4.4 ± 1.3 compared to 7.8 ± 0.5 for the uninfected control mice (P < 0.05). A similar fertility reduction was elicited in the wild-type (WT) C57BL/6 mice (4.3 ± 1.4 embryos/mouse) compared to the levels of the WT controls (9.1 ± 0.4 embryos/mouse) (P < 0.05). Following infection, WT mice mounted more robust humoral and cellular immune responses than HLA-DR4 mice. As determined by vaginal shedding, HLA-DR4 mice were more susceptible to a transcervical C. trachomatis D infection than WT mice. To assess if HLA-DR4 transgenic and WT mice could be protected by vaccination, 104 IFU of C. trachomatis D was delivered intranasally, and mice were challenged transcervically 6 weeks later with 5 × 105 IFU of C. trachomatis D. As determined by severity and length of vaginal shedding, WT C57BL/6 and HLA-DR4 mice were significantly protected by vaccination. The advantages and limitations of the HLA-DR4 transgenic mouse model for evaluating human C. trachomatis vaccine antigens are discussed.
Collapse
|
48
|
Banerjee A, Bhattacharya P, Dagur PK, Karmakar S, Ismail N, Joshi AB, Akue AD, KuKuruga M, McCoy JP, Dey R, Nakhasi HL. Live Attenuated Leishmania donovani Centrin Gene-Deleted Parasites Induce IL-23-Dependent IL-17-Protective Immune Response against Visceral Leishmaniasis in a Murine Model. THE JOURNAL OF IMMUNOLOGY 2017; 200:163-176. [PMID: 29187586 DOI: 10.4049/jimmunol.1700674] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022]
Abstract
No vaccine exists against visceral leishmaniasis. To develop effective vaccines, we have previously reported protective role of live attenuated centrin gene-deleted Leishmania donovani (LdCen-/- ) parasites through induction of Th1 type immune response in mice, hamsters, and dogs. In this study, we specifically explored the role of Th17 cells in LdCen-/- -induced host protection in mice. Our results showed that compared with wild-type L. donovani infection, LdCen-/- parasites induce significantly higher expression of Th17 differentiation cytokines in splenic dendritic cells. There was also induction of IL-17 and its promoting cytokines in total splenocytes and in both CD4 and CD8 T cells following immunization with LdCen-/- Upon challenge with wild-type parasites, IL-17 and its differentiating cytokines were significantly higher in LdCen-/- -immunized mice compared with nonimmunized mice that resulted in parasite control. Alongside IL-17 induction, we observed induction of IFN-γ-producing Th1 cells as reported earlier. However, Th17 cells are generated before Th1 cells. Neutralization of either IL-17 or IFN-γ abrogated LdCen-/- -induced host protection further confirming the essential role of Th17 along with Th1 cytokines in host protection. Treatment with recombinant IL-23, which is required for stabilization and maintenance of IL-17, heightened Th17, and Tc17 responses in immunized mice splenocytes. In contrast, Th17 response was absent in immunized IL-23R-/- mice that failed to induce protection upon virulent Leishmania challenge suggesting that IL-23 plays an essential role in IL-17-mediated protection by LdCen-/- parasites. This study unveiled the role of IL-23-dependent IL-17 induction in LdCen-/- parasite-induced immunity and subsequent protection against visceral leishmaniasis.
Collapse
Affiliation(s)
- Antara Banerjee
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993.,Department of Zoology, Bangabasi College, Kolkata, 700016 West Bengal, India
| | - Parna Bhattacharya
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Subir Karmakar
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Nevien Ismail
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Amritanshu B Joshi
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Adovi D Akue
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - Mark KuKuruga
- Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993
| | - John Philip McCoy
- Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Ranadhir Dey
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993;
| | - Hira L Nakhasi
- Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993;
| |
Collapse
|
49
|
Gomi R, Sharma A, Wu W, Sung B, Worgall S. Post-exposure immunization by capsid-modified AdC7 vector expressing Pseudomonas aeruginosa OprF clears P. aeruginosa respiratory infection. Vaccine 2017; 35:7174-7180. [PMID: 29126807 DOI: 10.1016/j.vaccine.2017.10.078] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 10/17/2017] [Accepted: 10/26/2017] [Indexed: 12/25/2022]
Abstract
Respiratory infections with Pseudomonas aeruginosa are major health problems, particularly in patients with cystic fibrosis (CF). No vaccine against P. aeruginosa is yet available. A vaccine that controls colonization of the respiratory tract with P. aeruginosa could be useful to prevent chronic infection and exacerbations. Replication-deficient adenoviral (Ad) vectors based on non-human serotypes are attractive vaccine platforms as they can circumvent the problem of pre-existing anti-Ad immunity in humans. The primate-based AdC7 vector AdC7OprF.RGD that expresses the outer membrane protein F (OprF) of P. aeruginosa (AdC7OprF) and that displays an integrin-binding arginine-glycine-aspartic acid (RGD) sequence is a potent inducer of lung mucosal and protective immunity. Here, we investigated the efficacy of immunization with AdC7OprF.RGD to clear an already established P. aeruginosa respiratory infection in mice (wild-type and CF) and rats. Intratracheal administration of the clinical P. aeruginosa strain RP73 embedded in agar beads was used to establish persistent infection. Subsequent intranasal immunization with AdC7OprF.RGD induced robust P. aeruginosa-specific systemic and mucosal, humoral and cellular immune responses. Importantly, the AdC7OprF.RGD immunized mice effectively cleared P. aeruginosa from the lungs. Likewise, immunization with AdC7OprF.RGD of CF mice and Sprague Dawley rats with established P. aeruginosa respiratory infection showed enhanced anti-Pseudomonas immune responses and increased clearance of P. aeruginosa from the lungs. These data suggest that AdC7OprF.RGD can be effective as a post-exposure vaccine and may be useful in clinical settings in particular for patients with CF who frequently harbor the bacteria over prolonged periods.
Collapse
Affiliation(s)
- Rika Gomi
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Anurag Sharma
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Wenzhu Wu
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Biin Sung
- Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Stefan Worgall
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States; Department of Genetic Medicine, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
50
|
Immunization with Bivalent Flagellin Protects Mice against Fatal Pseudomonas aeruginosa Pneumonia. J Immunol Res 2017; 2017:5689709. [PMID: 29201922 PMCID: PMC5671732 DOI: 10.1155/2017/5689709] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 08/25/2017] [Accepted: 09/10/2017] [Indexed: 01/21/2023] Open
Abstract
Pseudomonas aeruginosa lung infections present a major challenge to healthcare systems worldwide because they are commonly associated with high morbidity and mortality. Here, we demonstrate the protective efficacy of type a and b flagellins (bivalent flagellin) against acute fatal pneumonia in mice. Mice immunized intranasally with a bivalent flagellin vaccine were challenged by different flagellated strains of P. aeruginosa in an acute pneumonia model. Besides the protective effect of the vaccine, we further measured the host innate and cellular immunity responses. The immunized mice in our study were protected against both strains. Remarkably, active immunization with type a or b flagellin significantly improved survival of mice against heterologous strain compared to flagellin a or b antisera. We also showed that after an intranasal challenge by P. aeruginosa strain, neutrophils are recruited to the airways of vaccinated mice, and that the bivalent flagellin vaccine was proved to be protective by the generated CD4+IL-17+ Th17 cells. In conclusion, bivalent flagellin vaccine can confer protection against different strains of P. aeruginosa in an acute pneumonia mouse model by eliciting effective cellular and humoral immune responses, including increased IL-17 production and improved opsonophagocytic killing.
Collapse
|