1
|
Cuomo P, Medaglia C, Casillo A, Gentile A, Fruggiero C, Corsaro MM, Capparelli R. Phage-resistance alters Lipid A reactogenicity: a new strategy for LPS-based conjugate vaccines against Salmonella Rissen. Front Immunol 2024; 15:1450600. [PMID: 39723217 PMCID: PMC11668645 DOI: 10.3389/fimmu.2024.1450600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/22/2024] [Indexed: 12/28/2024] Open
Abstract
Salmonella enterica serovar Rissen (S. Rissen) is an emerging causative agent of foodborne diseases. The current emergence of antibiotic resistance makes necessary alternative therapeutic strategies. In this study, we investigated the potential of a phage-resistant strain of S. Rissen (RR) as a tool for developing an effective lipopolysaccharide (LPS)-based vaccine. The LPS O-antigen is known to play critical roles in protective immunity against Salmonella. However, the high toxicity of the LPS lipid A moiety limits its use in vaccines. Here, we demonstrated that the acquisition of bacteriophage resistance by S. Rissen leads to structural modifications in the LPS structure. Using NMR and mass spectrometry, we characterized the LPS from phage-resistant strains as a smooth variant bearing under-acylated Lipid A portions (penta- and tetra-acylated forms). We then combined RT-qPCR and NMR-based metabolomics to explore the effects of phage resistance and LPS modification on bacterial fitness and virulence. Finally, we conducted in vivo studies to determine whether lysogeny-induced remodeling of LPS affects the host immune response. Results revealed that the under-acylated variant of LPS from RR attenuates the inflammatory response in BALB/c mice, while eliciting a specific antibody response that protects against S. Rissen (RW) infection. In conclusion, our findings suggest that phage resistance, through lipid A modification, may offer a novel strategy for reducing LPS toxicity, highlighting its potential as a promising biological approach for developing LPS-based vaccines against Salmonella infections.
Collapse
Affiliation(s)
- Paola Cuomo
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Chiara Medaglia
- Functional Genomics Research Center, Fondazione Human Technopole, Milan, Italy
| | - Angela Casillo
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Antonio Gentile
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Carmine Fruggiero
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | | | - Rosanna Capparelli
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
2
|
Romerio A, Franco AR, Shadrick M, Shaik MM, Artusa V, Italia A, Lami F, Demchenko AV, Peri F. Overcoming Challenges in Chemical Glycosylation to Achieve Innovative Vaccine Adjuvants Possessing Enhanced TLR4 Activity. ACS OMEGA 2023; 8:36412-36417. [PMID: 37810727 PMCID: PMC10552098 DOI: 10.1021/acsomega.3c05363] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 08/23/2023] [Indexed: 10/10/2023]
Abstract
Lipopolysaccharide (LPS) mimicry leading to toll-like receptor 4 (TLR4) active compounds has been so far based mainly on reproducing the lipid A portion of LPS. Our work led to a series of structurally simplified synthetic TLR4 agonists in preclinical development as vaccine adjuvants called FPs. FPs bind MD2/TLR4 similarly to lipid A, inserting the lipid chains in the MD2 lipophilic cavity. A strategy to improve FPs' target affinity is introducing a monosaccharide unit in C6, mimicking the first sugar of the LPS core. We therefore designed a panel of FP derivatives bearing different monosaccharides in C6. We report here the synthesis and optimization of FPs' C6 glycosylation, which presented unique challenges and limitations. The biological activity of glycosylated FP compounds was preliminarily assessed in vitro in HEK-Blue cells. The new molecules showed a higher potency in stimulating TLR4 activation when compared to the parent molecule while maintaining TLR4 selectivity.
Collapse
Affiliation(s)
- Alessio Romerio
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Ana Rita Franco
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Melanie Shadrick
- Department
of Chemistry, Saint Louis University, 3501 Laclede Avenue, St. Louis, Missouri 63103, United States
| | - Mohammed Monsoor Shaik
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Valentina Artusa
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Alice Italia
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Federico Lami
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Alexei V. Demchenko
- Department
of Chemistry, Saint Louis University, 3501 Laclede Avenue, St. Louis, Missouri 63103, United States
| | - Francesco Peri
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| |
Collapse
|
3
|
Firmal P, Shah VK, Pant R, Chattopadhyay S. RING finger protein TOPORS modulates the expression of tumor suppressor SMAR1 in colorectal cancer via the TLR4-TRIF pathway. Mol Oncol 2021; 16:1523-1540. [PMID: 34689394 PMCID: PMC8978522 DOI: 10.1002/1878-0261.13126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
TOP1‐binding arginine/serine‐rich protein (TOPORS), a really interesting new gene finger protein, has the ability to bind to a palindromic consensus DNA sequence that enables it to function as a potential transcriptional regulator. However, its role in regulating the transcription of cancer‐associated genes is yet to be explored. As Toll‐like receptor 4 (TLR4) agonists are known to regress solid tumors, we observed that lipopolysaccharide (LPS) induces TOPORS via a TLR4‐TIR domain‐containing adapter‐inducing interferon‐β‐dependent pathway, which in turn modulates the transcription of tumor suppressor scaffold/matrix attachment region‐binding protein 1 (SMAR1, also known as BANP). ChIP analysis showed that TOPORS binds on the SMAR1 promoter and its occupancy increases upon LPS treatment. A previous study from our laboratory revealed that SMAR1 acts as a repressor of signal transducer and activator of transcription 3 (STAT3) transcription. Tumor growth, as well as tumor‐associated macrophage polarization, depends on the status of the STAT1:STAT3 ratio. LPS‐induced SMAR1 expression decreases STAT3 expression and also skews the macrophage polarization toward M1 phenotype. In contrast, LPS failed to polarize tumor‐associated macrophages to M1 phenotype in a SMAR1‐silenced condition, which shows the involvement of SMAR1 in dictating the fate of colorectal cancer progression. Identification of the molecular mechanism behind LPS‐mediated tumor regression would be crucial for designing cancer treatment strategies involving bacterial components.
Collapse
Affiliation(s)
- Priyanka Firmal
- National Centre for Cell Science, S.P. Pune University Campus, Ganeshkhind, Pune, 411007
| | - Vibhuti Kumar Shah
- National Centre for Cell Science, S.P. Pune University Campus, Ganeshkhind, Pune, 411007
| | - Richa Pant
- National Centre for Cell Science, S.P. Pune University Campus, Ganeshkhind, Pune, 411007
| | - Samit Chattopadhyay
- National Centre for Cell Science, S.P. Pune University Campus, Ganeshkhind, Pune, 411007.,Department of Biological Sciences, BITS Pilani, K. K. Birla Goa Campus, NH 17B, Zuarinagar, Goa, 403726.,Indian Institute of Chemical Biology, Raja S C Mullick Road, Jadavpur, Kolkata, 700032
| |
Collapse
|
4
|
Facchini F, Minotti A, Luraghi A, Romerio A, Gotri N, Matamoros-Recio A, Iannucci A, Palmer C, Wang G, Ingram R, Martin-Santamaria S, Pirianov G, De Andrea M, Valvano MA, Peri F. Synthetic Glycolipids as Molecular Vaccine Adjuvants: Mechanism of Action in Human Cells and In Vivo Activity. J Med Chem 2021; 64:12261-12272. [PMID: 34382796 PMCID: PMC8404200 DOI: 10.1021/acs.jmedchem.1c00896] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Indexed: 01/07/2023]
Abstract
Modern adjuvants for vaccine formulations are immunostimulating agents whose action is based on the activation of pattern recognition receptors (PRRs) by well-defined ligands to boost innate and adaptive immune responses. Monophosphoryl lipid A (MPLA), a detoxified analogue of lipid A, is a clinically approved adjuvant that stimulates toll-like receptor 4 (TLR4). The synthesis of MPLA poses manufacturing and quality assessment challenges. Bridging this gap, we report here the development and preclinical testing of chemically simplified TLR4 agonists that could sustainably be produced in high purity and on a large scale. Underpinned by computational and biological experiments, we show that synthetic monosaccharide-based molecules (FP compounds) bind to the TLR4/MD-2 dimer with submicromolar affinities stabilizing the active receptor conformation. This results in the activation of MyD88- and TRIF-dependent TLR4 signaling and the NLRP3 inflammasome. FP compounds lack in vivo toxicity and exhibit adjuvant activity by stimulating antibody responses with a potency comparable to MPLA.
Collapse
Affiliation(s)
- Fabio
A. Facchini
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Alberto Minotti
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Andrea Luraghi
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Alessio Romerio
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Nicole Gotri
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| | - Alejandra Matamoros-Recio
- Department
of Structural and Chemical Biology, Centro
de Investigaciones Biologicas Margarita Salas, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Andrea Iannucci
- Department
of Translational Medicine, University of
Eastern Piedmont, 28100 Novara, Italy
- CAAD—Center
for Translational Research on Autoimmune and Allergic Disease, University of Eastern Piedmont, 28100 Novara, Italy
| | - Charys Palmer
- Department
of Biomedical and Forensic Sciences, Anglia
Ruskin University, East Road, Cambridge CB1
1PT, U.K.
| | - Guanbo Wang
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University of Belfast; 97 Lisburn Road, Belfast BT9 7BL, U.K.
| | - Rebecca Ingram
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University of Belfast; 97 Lisburn Road, Belfast BT9 7BL, U.K.
| | - Sonsoles Martin-Santamaria
- Department
of Structural and Chemical Biology, Centro
de Investigaciones Biologicas Margarita Salas, C/Ramiro de Maeztu, 9, 28040 Madrid, Spain
| | - Grisha Pirianov
- Department
of Biomedical and Forensic Sciences, Anglia
Ruskin University, East Road, Cambridge CB1
1PT, U.K.
| | - Marco De Andrea
- CAAD—Center
for Translational Research on Autoimmune and Allergic Disease, University of Eastern Piedmont, 28100 Novara, Italy
- Department
of Public Health and Pediatric Sciences, University of Turin, Medical School, 10126 Turin, Italy
| | - Miguel A. Valvano
- The Wellcome-Wolfson
Institute for Experimental Medicine, Queen’s
University of Belfast; 97 Lisburn Road, Belfast BT9 7BL, U.K.
| | - Francesco Peri
- Department
of Biotechnology and Biosciences, University
of Milano-Bicocca, Piazza della Scienza, 2, 20126 Milano, Italy
| |
Collapse
|
5
|
Wu D, Fan Z, Li J, Zhang Y, Wang C, Xu Q, Wang L. Evaluation of Alpha-Ketoglutarate Supplementation on the Improvement of Intestinal Antioxidant Capacity and Immune Response in Songpu Mirror Carp ( Cyprinus carpio) After Infection With Aeromonas hydrophila. Front Immunol 2021; 12:690234. [PMID: 34220849 PMCID: PMC8250152 DOI: 10.3389/fimmu.2021.690234] [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: 04/02/2021] [Accepted: 06/04/2021] [Indexed: 11/24/2022] Open
Abstract
As an intermediate substance of the tricarboxylic acid cycle and a precursor substance of glutamic acid synthesis, the effect of alpha-ketoglutarate on growth and protein synthesis has been extensively studied. However, its prevention and treatment of pathogenic bacteria and its mechanism have not yet been noticed. To evaluate the effects of alpha-ketoglutarate on intestinal antioxidant capacity and immune response of Songpu mirror carp, a total of 360 fish with an average initial weight of 6.54 ± 0.08 g were fed diets containing alpha-ketoglutarate with 1% for 8 weeks. At the end of the feeding trial, the fish were challenged with Aeromonas hydrophila for 2 weeks. The results indicated that alpha-ketoglutarate supplementation significantly increased the survival rate of carp after infection with Aeromonas hydrophila (P < 0.05), and the contents of immune digestion enzymes including lysozyme, alkaline phosphatase and the concentration of complement C4 were markedly enhanced after alpha-ketoglutarate supplementation (P < 0.05). Also, appropriate alpha-ketoglutarate increased the activities of total antioxidant capacity and catalase and prevented the up-regulation in the mRNA expression levels of pro-inflammatory cytokines including tumor necrosis factor-α, interleukin-1β, interleukin-6, and interleukin-8 (P < 0.05). Furthermore, the mRNA expression levels of toll-like receptor 4 (TLR4), and nuclear factor kappa-B (NF-κB) were strikingly increased after infection with Aeromonas hydrophila (P < 0.05), while the TLR4 was strikingly decreased with alpha-ketoglutarate supplementation (P < 0.05). Moreover, the mRNA expression levels of tight junctions including claudin-1, claudin-3, claudin-7, claudin-11 and myosin light chain kinases (MLCK) were upregulated after alpha-ketoglutarate supplementation (P < 0.05). In summary, the appropriate alpha-ketoglutarate supplementation could increase survival rate, strengthen the intestinal enzyme immunosuppressive activities, antioxidant capacities and alleviate the intestinal inflammation, thereby promoting the intestinal immune responses and barrier functions of Songpu mirror carp via activating TLR4/MyD88/NF-κB and MLCK signaling pathways after infection with Aeromonas hydrophila.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Ze Fan
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Jinnan Li
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Yuanyuan Zhang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Chang'an Wang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| | - Qiyou Xu
- School of Life Science, Huzhou University, Huzhou, China
| | - Liansheng Wang
- Key Laboratory of Aquatic Animal Diseases and Immune Technology of Heilongjiang Province, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin, China
| |
Collapse
|
6
|
Effect of lipopolysaccharide structure on functional response of whole blood cells. Immunobiology 2020; 226:152030. [PMID: 33278708 DOI: 10.1016/j.imbio.2020.152030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/06/2020] [Accepted: 11/18/2020] [Indexed: 01/10/2023]
Abstract
Lipopolysaccharides (LPSs) induce a wide spectrum of functional activities after interaction with blood cells. Effect of structure of toxic LPS from S- and Re-chemotypes of E. coli and/or non-toxic LPS of Rhodobacter capsulatus PG (R. caps.) on activation of neutrophils and monocytes of human whole blood were studied, particularly, expression of TLR4, CD14 and CD11b receptors, phagocytosis of BioParticles Alexa Fluor 488, synthesis of cytokines and chemokines. A leading role of CD11b receptor in phagocytic activity of neutrophils primed by LPS from various E. coli chemotypes was shown. The non-toxic LPS of R. caps. does not affect the efficiency of phagocytosis activity of the neutrophils. The LPS of R. caps. was shown to induce production of TRIF-dependent cytokine IFN-β in human whole blood leukocytes selectively, without activating MyD88-dependent pathway of pro-inflammatory cytokine synthesis, displaying properties of patrial agonist of TLR4. Structure and biological activity of LPS R. caps. allows considering it as a promising immunity stimulating pharmacological agent.
Collapse
|
7
|
Δ42PD1-TLR4 Augments γδ-T Cell Activation of the Transitional Memory Subset of CD4 + T Cells. iScience 2020; 23:101620. [PMID: 33089108 PMCID: PMC7567942 DOI: 10.1016/j.isci.2020.101620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/25/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
TLR ligands can contribute to T cell immune responses by indirectly stimulating antigen presentation and cytokines and directly serving as co-stimulatory signals. We have previously reported that the human endogenous surface protein, Δ42PD1, is expressed primarily on (Vγ9)Vδ2 cells and can interact with TLR4. Since Vδ2 cells possess antigen presentation capacity, we sought to further characterize if the Δ42PD1-TLR4 interaction has a role in stimulating T cell responses. In this study, we found that stimulation of Vδ2 cells not only upregulated Δ42PD1 expression but also increased MHC class II molecules necessary for the antigen presentation. In a mixed leukocyte reaction assay, upregulation of Δ42PD1 on Vδ2 cells elevated subsequent T cell proliferation. Furthermore, the interaction between Δ42PD1-TLR4 augments Vδ2 cell stimulation of autologous CMV pp65-or TT-specific CD4+ T cell proliferation and IFN-γ responses, which was specifically and significantly reduced by blocking the Δ42PD1-TLR4 interaction. Furthermore, confocal microscopy analysis confirmed the interaction between Δ42PD1+HLA-DR+Vδ2 cells and TLR4+CD4 T cells. Interestingly, the subset of CD4+ T cells expressing TLR4 appears to be PD-1+ CD45RO+CD45RA+ transitional memory T cells and responded to Δ42PD1+HLA-DR+Vδ2 cells. Overall, this study demonstrated an important biological role of Δ42PD1 protein exhibited by Vδ2 antigen-presenting cells in augmenting T cell activation through TLR4, which may serve as an additional co-stimulatory signal. Δ42PD1 is co-expressed with MHC-II on activated Vδ2 cells Δ42PD1+MHC-II+Vδ2 cells interact directly with TLR4+CD4+T cells in 3D imaging TLR4 is highly expressed on the PD-1+CD45RO+CD45RA+CD4+T cell subset Δ42PD1-TLR4 selectively activates this subset of Ag-specific CD4+ T cells
Collapse
|
8
|
Vemireddy S, Madhurantakam PP, Talati MN, Sampath Kumar HM. Cationic pH-Responsive Polycaprolactone Nanoparticles as Intranasal Antigen Delivery System for Potent Humoral and Cellular Immunity against Recombinant Tetravalent Dengue Antigen. ACS APPLIED BIO MATERIALS 2019; 2:4837-4846. [DOI: 10.1021/acsabm.9b00597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Sravanthi Vemireddy
- Vaccine Immunology Laboratory, OSPC Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, CSIR−Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Preethi Pallavi Madhurantakam
- Vaccine Immunology Laboratory, OSPC Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, CSIR−Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Mamta N Talati
- Vaccine Immunology Laboratory, OSPC Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, CSIR−Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Halmuthur M. Sampath Kumar
- Vaccine Immunology Laboratory, OSPC Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, CSIR−Indian Institute of Chemical Technology, Hyderabad 500007, India
| |
Collapse
|
9
|
Teixeira AF, Fernandes LG, Cavenague MF, Takahashi MB, Santos JC, Passalia FJ, Daroz BB, Kochi LT, Vieira ML, Nascimento AL. Adjuvanted leptospiral vaccines: Challenges and future development of new leptospirosis vaccines. Vaccine 2019; 37:3961-3973. [DOI: 10.1016/j.vaccine.2019.05.087] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/16/2019] [Accepted: 05/21/2019] [Indexed: 12/26/2022]
|
10
|
Apostólico JDS, Lunardelli VAS, Yamamoto MM, Cunha-Neto E, Boscardin SB, Rosa DS. Poly(I:C) Potentiates T Cell Immunity to a Dendritic Cell Targeted HIV-Multiepitope Vaccine. Front Immunol 2019; 10:843. [PMID: 31105693 PMCID: PMC6492566 DOI: 10.3389/fimmu.2019.00843] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 04/01/2019] [Indexed: 02/05/2023] Open
Abstract
Cellular immune responses are implicated in resistance to HIV and have been considered for the development of an effective vaccine. Despite their safety profile, subunit vaccines need to be delivered combined with an adjuvant. In the last years, in vivo antigen targeting to dendritic cells (DCs) using chimeric monoclonal antibodies (mAb) against the DC endocytic receptor DEC205/CD205 was shown to support long-term T cell immunity. Here, we evaluated the ability of different adjuvants to modulate specific cellular immune response when eight CD4+ HIV-derived epitopes (HIVBr8) were targeted to DEC205+ DCs in vivo. Immunization with two doses of αDECHIVBr8 mAb along with poly(I:C) induced Th1 cytokine production and higher frequency of HIV-specific polyfunctional and long-lived T cells than MPL or CpG ODN-assisted immunization. Although each adjuvant elicited responses against the 8 epitopes present in the vaccine, the magnitude of the T cell response was higher in the presence of poly(I:C). Moreover, poly(I:C) up regulated the expression of costimulatory molecules in both cDC1 and cDC2 DCs subsets. In summary, the use of poly(I:C) in a vaccine formulation that targets multiple epitopes to the DEC205 receptor improved the potency and the quality of HIV-specific responses when compared to other vaccine-adjuvant formulations. This study highlights the importance of the rational selection of antigen/adjuvant combination to potentiate the desired immune responses.
Collapse
Affiliation(s)
- Juliana de Souza Apostólico
- Laboratory of Experimental Vaccines, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii)-INCT, São Paulo, Brazil
| | - Victória Alves Santos Lunardelli
- Laboratory of Experimental Vaccines, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii)-INCT, São Paulo, Brazil
| | - Marcio Massao Yamamoto
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Institute for Investigation in Immunology (iii)-INCT, São Paulo, Brazil.,Laboratory of Clinical Immunology and Allergy (LIM60), School of Medicine-University of São Paulo, São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Institute for Investigation in Immunology (iii)-INCT, São Paulo, Brazil.,Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniela Santoro Rosa
- Laboratory of Experimental Vaccines, Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil.,Institute for Investigation in Immunology (iii)-INCT, São Paulo, Brazil
| |
Collapse
|
11
|
Mosaheb M, Wetzler LM. Meningococcal PorB induces a robust and diverse antigen specific T cell response as a vaccine adjuvant. Vaccine 2018; 36:7689-7699. [PMID: 30381152 DOI: 10.1016/j.vaccine.2018.10.074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/02/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
Vaccines formulated with adjuvant have been effective against numerous infectious diseases, almost always due to induction of functional antibodies that recognizes the pathogen of interest. There is an unmet clinical need for vaccine adjuvants that induce T cells responses to potentially enhance protection against malignancies and intracellular pathogens, where a humoral response, alone, may not be adequate for protection. In this study, we demonstrate that a TLR2 ligand-based adjuvant, meningococcal PorB, has broad immunostimulatory activity with the ability to induce a robust and diverse vaccine antigen specific T cell response. We demonstrate that a vaccine formulated with PorB admixed with ovalbumin induces a wide variety of antigen specific antibody subclasses and effector molecules (MIG, MCP-1, IP-10, MIP-1α, KC & IL-2) with known roles for inducing T cell responses, along with elevated levels of Th1 and Th2 type cytokines upon antigen stimulation. We confirmed production of these cytokines by examining the antigen-specific T cells induced by PorB in vivo. After two immunizations with vaccine formulated with PorB/OVA, antigen-specific CD4 and CD8 T cells were significantly increased in numbers and produced IL-4 or IFN-γ upon ex vivo antigen re-stimulation. Finally, in a Listeria mouse infection model, vaccine formulated with PorB significantly reduced the bacterial burden upon a low dose infection and increased survival upon a high dose infection with recombinant Listeria monocytogenes engineered to express OVA (rLmOVA), a pathogen that requires OVA-antigen specific cytotoxic CD8 T cells for clearance. In summary, PorB is able to induce antigen specific broad B and T cell responses, illustrating its potential as a potent and new vaccine adjuvant.
Collapse
Affiliation(s)
- Munir Mosaheb
- Dept. of Microbiology, Boston University School of Medicine, USA
| | - Lee M Wetzler
- Dept. of Microbiology, Boston University School of Medicine, USA; Dept. of Medicine, Sect. of Infectious Diseases, Boston Medical Center, Boston, MA 02118, USA.
| |
Collapse
|
12
|
Watts BA, George T, Sherwood ER, Good DW. Monophosphoryl lipid A prevents impairment of medullary thick ascending limb [Formula: see text] absorption and improves plasma [Formula: see text] concentration in septic mice. Am J Physiol Renal Physiol 2018; 315:F711-F725. [PMID: 29741098 PMCID: PMC6172583 DOI: 10.1152/ajprenal.00033.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/10/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023] Open
Abstract
Metabolic acidosis is the most common acid-base disorder in septic patients and is associated with increased mortality. Previously, we demonstrated that sepsis induced by cecal ligation and puncture (CLP) impairs [Formula: see text] absorption in the medullary thick ascending limb (MTAL) by 1) decreasing the intrinsic [Formula: see text] absorptive capacity and 2) enhancing inhibition of [Formula: see text] absorption by LPS through upregulation of Toll-like receptor (TLR) 4 signaling. Both effects depend on ERK activation. Monophosphoryl lipid A (MPLA) is a detoxified TLR4 agonist that enhances innate antimicrobial immunity and improves survival following sepsis. Pretreatment of MTALs with MPLA in vitro prevents LPS inhibition of [Formula: see text] absorption. Here we examined whether pretreatment with MPLA would protect the MTAL against sepsis. Vehicle or MPLA was administered to mice 48 h before sham or CLP surgery, and MTALs were studied in vitro 18 h postsurgery. Pretreatment with MPLA prevented the effects of sepsis to decrease the basal [Formula: see text] absorption rate and enhance inhibition by LPS. These protective effects were mediated through MPLA stimulation of a Toll/IL-1 receptor domain-containing adaptor-inducing IFN-β-(TRIF)-dependent phosphatidylinositol 3-kinase-Akt pathway that prevents sepsis- and LPS-induced ERK activation. The effects of MPLA to improve MTAL [Formula: see text] absorption were associated with marked improvement in plasma [Formula: see text] concentration, supporting a role for the kidneys in the pathogenesis of sepsis-induced metabolic acidosis. These studies support detoxified TLR4-based immunomodulators, such as MPLA, that enhance antimicrobial responses as a safe and effective approach to prevent or treat sepsis-induced renal tubule dysfunction and identify cell signaling pathways that can be targeted to preserve MTAL [Formula: see text] absorption and attenuate metabolic acidosis during sepsis.
Collapse
Affiliation(s)
- Bruns A Watts
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Thampi George
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David W Good
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
13
|
Targeting pattern-recognition receptors to discover new small molecule immune modulators. Eur J Med Chem 2018; 144:82-92. [DOI: 10.1016/j.ejmech.2017.12.026] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/03/2017] [Accepted: 12/07/2017] [Indexed: 12/21/2022]
|
14
|
Abstract
Adjuvants have been deliberately added to vaccines since the 1920's when alum was discovered to boost antibody responses, leading to better protection. The first adjuvants were discovered by accident and were used in the safer but less immunogenic subunit vaccines, supposedly by providing an antigen depot to extend antigen presentation. Since that time, much has been discovered about how these adjuvants impact cells at the tissue site to activate innate immune responses, mobilize dendritic cells and drive adaptive immunity. New approaches to vaccine construction for infectious diseases that have so far not been well addressed by conventional vaccines often attempt to induce antibodies, polyfunctional CD4+ T cells and CD8+ CTLs. The discovery of pattern recognition receptors and ligands that drive desired T cell responses has led to development of novel adjuvant strategies using immunomodulatory agents to direct appropriate immune responses.
Collapse
Affiliation(s)
- Amy S McKee
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| | - Philippa Marrack
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Biomedical Research, National Jewish Health, 1400, Jackson St., Denver, CO 80206, USA
| |
Collapse
|
15
|
Shinde P, Liu W, Ménoret A, Luster AD, Vella AT. Optimal CD4 T cell priming after LPS-based adjuvanticity with CD134 costimulation relies on CXCL9 production. J Leukoc Biol 2017; 102:57-69. [PMID: 28432083 DOI: 10.1189/jlb.1a0616-261rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 03/29/2017] [Accepted: 04/02/2017] [Indexed: 12/29/2022] Open
Abstract
LPS is a powerful adjuvant, and although LPS-mediated TLR4 signaling has been exquisitely delineated, the in vivo mechanism of how TLR4 responses impact T cell priming is far less clear. Besides costimulation, TNF and type 1 IFN are dominant cytokines released after TLR4 activation and can shape T cell responses, but other downstream factors have not been examined extensively. Depending on context, we show that IFNαR1 blockade resulted in minor to major effects on specific CD4 T cell clonal expansion. To help explain these differences, it was hypothesized that IFNαR1 blockade would inhibit specific T cell migration by reducing chemokine receptor signaling, but specific CD4 T cells from IFNαR1-blocked mice were readily able to migrate in response to specific chemokines. Next, we examined downstream factors and found that type 1 IFN signaling was necessary for chemokine production, even when mice were immunized with specific Ag with LPS and CD134 costimulation. IFNαR1 signaling promoted CXCL9 and CXCL10 synthesis, suggesting that these chemokines might be involved in the LPS and CD134 costimulation response. After immunization, we show that CXCL9 blockade inhibited CD4 T cell accumulation in the liver but also in LNs, even in the presence of elevated serum IFN-β levels. Thus, whereas type 1 IFN might have direct effects on primed CD4 T cells, the downstream chemokines that play a role during migration also impact accumulation. In sum, CXCL9 production is a key benchmark for productive CD4 T cell vaccination strategies.
Collapse
Affiliation(s)
- Paurvi Shinde
- Department of Immunology, School of Medicine, University of Connecticut Health, Farmington, Connecticut, USA
| | - Wenhai Liu
- Department of Immunology, School of Medicine, University of Connecticut Health, Farmington, Connecticut, USA
| | - Antoine Ménoret
- Department of Immunology, School of Medicine, University of Connecticut Health, Farmington, Connecticut, USA.,Institute for Systems Genomics, University of Connecticut School of Medicine, Farmington, Connecticut, USA; and
| | - Andrew D Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anthony T Vella
- Department of Immunology, School of Medicine, University of Connecticut Health, Farmington, Connecticut, USA;
| |
Collapse
|
16
|
Li HR, Liu J, Zhang SL, Luo T, Wu F, Dong JH, Guo YJ, Zhao L. Corilagin ameliorates the extreme inflammatory status in sepsis through TLR4 signaling pathways. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:18. [PMID: 28056977 PMCID: PMC5217594 DOI: 10.1186/s12906-016-1533-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 12/19/2016] [Indexed: 11/18/2022]
Abstract
Background Sepsis is one of the serious disorders in clinical practice. Recent studies found toll-like receptors 4 (TLR4) played an important role in sepsis. In this study, we tried to find the influence of Corilagin on TLR4 signal pathways in vitro and in vivo. Methods The cellular and animal models of sepsis were established by LPS and then interfered with Corilagin. Real-time PCR and western blot were employed to detect the mRNA and protein expressions of TLR4, MyD88, TRIF and TRAF6. ELISA was used to determine the IL-6 and IL-1β levels in supernatant and serum. Results The survival rate was improved in the LPS + Corilagin group, and the mRNA and protein expressions of TLR4, MyD88, TRIF and TRAF6 were significantly decreased than that in the LPS group both in cellular and animal models (P < 0.01). The pro-inflammatory cytokines IL-6 and IL-1β were greatly decreased in the LPS + Corilagin group both in supernatant and serum (P < 0.01). Conclusions Corilagin exerts the anti-inflammatory effects by down-regulating the TLR4 signaling molecules to ameliorate the extreme inflammatory status in sepsis.
Collapse
|
17
|
Zariri A, Pupo E, van Riet E, van Putten JPM, van der Ley P. Modulating endotoxin activity by combinatorial bioengineering of meningococcal lipopolysaccharide. Sci Rep 2016; 6:36575. [PMID: 27841285 PMCID: PMC5107901 DOI: 10.1038/srep36575] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/10/2016] [Indexed: 11/16/2022] Open
Abstract
Neisseria meningitidis contains a very potent hexa-acylated LPS that is too toxic for therapeutic applications. We used systematic molecular bioengineering of meningococcal LPS through deletion of biosynthetic enzymes in combination with induction of LPS modifying enzymes to yield a variety of novel LPS mutants with changes in both lipid A acylation and phosphorylation. Mass spectrometry was used for detailed compositional determination of the LPS molecular species, and stimulation of immune cells was done to correlate this with endotoxic activity. Removal of phosphethanolamine in lipid A by deletion of lptA slightly reduces activity of hexa-acylated LPS, but this reduction is even more evident in penta-acylated LPS. Surprisingly, expression of PagL deacylase in a penta-acylated lpxL1 mutant increased LPS activity, contradicting the general rule that tetra-acylated LPS is less active than penta-acylated LPS. Further modification included expression of lpxP, an enzyme known to add a secondary 9-hexadecenoic acid to the 2’ acyl chain. The LpxP enzyme is temperature-sensitive, enabling control over the ratio of expressed modified hexa- and penta-acylated LPS by simply changing the growth temperature. These LPS derivatives display a broad range of TLR4 activity and differential cytokine induction, which can be exploited for use as vaccine adjuvant or other TLR4-based therapeutics.
Collapse
Affiliation(s)
- Afshin Zariri
- Institute for Translational Vaccinology (Intravacc), Antonie van Leeuwenhoeklaan 9, 3720 AL Bilthoven, the Netherlands.,Department of Infectious Diseases and Immunology, Utrecht University, 3584 CL, Utrecht, the Netherlands
| | - Elder Pupo
- Institute for Translational Vaccinology (Intravacc), Antonie van Leeuwenhoeklaan 9, 3720 AL Bilthoven, the Netherlands
| | - Elly van Riet
- Institute for Translational Vaccinology (Intravacc), Antonie van Leeuwenhoeklaan 9, 3720 AL Bilthoven, the Netherlands
| | - Jos P M van Putten
- Department of Infectious Diseases and Immunology, Utrecht University, 3584 CL, Utrecht, the Netherlands
| | - Peter van der Ley
- Institute for Translational Vaccinology (Intravacc), Antonie van Leeuwenhoeklaan 9, 3720 AL Bilthoven, the Netherlands
| |
Collapse
|
18
|
Hernandez A, Bohannon JK, Luan L, Fensterheim BA, Guo Y, Patil NK, McAdams C, Wang J, Sherwood ER. The role of MyD88- and TRIF-dependent signaling in monophosphoryl lipid A-induced expansion and recruitment of innate immunocytes. J Leukoc Biol 2016; 100:1311-1322. [PMID: 27354411 DOI: 10.1189/jlb.1a0216-072r] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/01/2016] [Accepted: 06/12/2016] [Indexed: 12/26/2022] Open
Abstract
Treatment with the TLR4 agonist MPLA augments innate resistance to common bacterial pathogens. However, the cellular and molecular mechanisms by which MPLA augments innate immunocyte functions are not well characterized. This study examined the importance of MyD88- and TRIF-dependent signaling for leukocyte mobilization, recruitment, and activation following administration of MPLA. MPLA potently induced MyD88- and TRIF-dependent signaling. A single injection of MPLA caused rapid mobilization and recruitment of neutrophils, a response that was largely mediated by the chemokines CXCL1 and -2 and the hemopoietic factor G-CSF. Rapid neutrophil recruitment and chemokine production were regulated by both pathways although the MyD88-dependent pathway showed some predominance. In further studies, multiple injections of MPLA potently induced mobilization and recruitment of neutrophils and monocytes. Neutrophil recruitment after multiple injections of MPLA was reliant on MyD88-dependent signaling, but effective monocyte recruitment required activation of both pathways. MPLA treatment induced expansion of myeloid progenitors in bone marrow and upregulation of CD11b and shedding of L-selectin by neutrophils, all of which were attenuated in MyD88- and TRIF-deficient mice. These results show that MPLA-induced neutrophil and monocyte recruitment, expansion of bone marrow progenitors and augmentation of neutrophil adhesion molecule expression are regulated by both the MyD88- and TRIF-dependent pathways.
Collapse
Affiliation(s)
- Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Liming Luan
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Benjamin A Fensterheim
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Yin Guo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Naeem K Patil
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Chase McAdams
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jingbin Wang
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee; and
| |
Collapse
|
19
|
Pereira FV, Melo ACL, de Melo FM, Mourão-Sá D, Silva P, Berzaghi R, Herbozo CCA, Coelho-Dos-Reis J, Scutti JA, Origassa CST, Pereira RM, Juliano L, Juliano MA, Carmona AK, Câmara NOS, Tsuji M, Travassos LR, Rodrigues EG. TLR4-mediated immunomodulatory properties of the bacterial metalloprotease arazyme in preclinical tumor models. Oncoimmunology 2016; 5:e1178420. [PMID: 27622031 DOI: 10.1080/2162402x.2016.1178420] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 04/07/2016] [Accepted: 04/10/2016] [Indexed: 02/09/2023] Open
Abstract
Despite the recent approval of new agents for metastatic melanoma, its treatment remains challenging. Moreover, few available immunotherapies induce a strong cellular immune response, and selection of the correct immunoadjuvant is crucial for overcoming this obstacle. Here, we studied the immunomodulatory properties of arazyme, a bacterial metalloprotease, which was previously shown to control metastasis in a murine melanoma B16F10-Nex2 model. The antitumor activity of arazyme was independent of its proteolytic activity, since heat-inactivated protease showed comparable properties to the active enzyme; however, the effect was dependent on an intact immune system, as antitumor properties were lost in immunodeficient mice. The protective response was IFNγ-dependent, and CD8(+) T lymphocytes were the main effector antitumor population, although B and CD4(+) T lymphocytes were also induced. Macrophages and dendritic cells were involved in the induction of the antitumor response, as arazyme activation of these cells increased both the expression of surface activation markers and proinflammatory cytokine secretion through TLR4-MyD88-TRIF-dependent, but also MAPK-dependent pathways. Arazyme was also effective in the murine breast adenocarcinoma 4T1 model, reducing primary and metastatic tumor development, and prolonging survival. To our knowledge, this is the first report of a bacterial metalloprotease interaction with TLR4 and subsequent receptor activation that promotes a proinflammatory and tumor protective response. Our results show that arazyme has immunomodulatory properties, and could be a promising novel alternative for metastatic melanoma treatment.
Collapse
Affiliation(s)
- Felipe V Pereira
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP), São Paulo, Brazil; Department of Immunology, Instituto de Ciências Biomédicas (ICB), University of São Paulo (USP), São Paulo, Brazil; HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center (ADARC), Rockefeller University, NY, USA
| | - Amanda C L Melo
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP), São Paulo, Brazil; Department of Immunology, Instituto de Ciências Biomédicas (ICB), University of São Paulo (USP), São Paulo, Brazil
| | - Filipe M de Melo
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Diego Mourão-Sá
- Immunobiology Laboratory, Cancer Research UK, London Research Institute , London, UK
| | - Priscila Silva
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Rodrigo Berzaghi
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Carolina C A Herbozo
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Jordana Coelho-Dos-Reis
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center (ADARC), Rockefeller University, NY, USA; Rene Rachou Research Center, Oswaldo Cruz Foundation, FIOCRUZ, Minas Gerais, Brazil
| | - Jorge A Scutti
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Clarice S T Origassa
- Department of Immunology, Instituto de Ciências Biomédicas (ICB), University of São Paulo (USP) , São Paulo, Brazil
| | - Rosana M Pereira
- Department of Immunology, Instituto de Ciências Biomédicas (ICB), University of São Paulo (USP) , São Paulo, Brazil
| | - Luis Juliano
- Department of Biophysics, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Maria Aparecida Juliano
- Department of Biophysics, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Adriana K Carmona
- Department of Biophysics, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Niels O S Câmara
- Department of Immunology, Instituto de Ciências Biomédicas (ICB), University of São Paulo (USP) , São Paulo, Brazil
| | - Moriya Tsuji
- HIV and Malaria Vaccine Program, Aaron Diamond AIDS Research Center (ADARC), Rockefeller University , NY, USA
| | - Luiz R Travassos
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| | - Elaine G Rodrigues
- Department of Microbiology, Immunology, and Parasitology, Escola Paulista de Medicina (EPM), Federal University of São Paulo (UNIFESP) , São Paulo, Brazil
| |
Collapse
|
20
|
Biedroń R, Peruń A, Józefowski S. CD36 Differently Regulates Macrophage Responses to Smooth and Rough Lipopolysaccharide. PLoS One 2016; 11:e0153558. [PMID: 27073833 PMCID: PMC4830570 DOI: 10.1371/journal.pone.0153558] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 03/31/2016] [Indexed: 01/22/2023] Open
Abstract
Lipopolysaccharide (LPS) is the major pathogen-associated molecular pattern of Gram-negative bacterial infections, and includes smooth (S-LPS) and rough (R-LPS) chemotypes. Upon activation by LPS through CD14, TLR4/MD-2 heterodimers sequentially induce two waves of intracellular signaling for macrophage activation: the MyD88-dependent pathway from the plasma membrane and, following internalization, the TRIF-dependent pathway from endosomes. We sought to better define the role of scavenger receptors CD36 and CD204/SR-A as accessory LPS receptors that can contribute to pro-inflammatory and microbicidal activation of macrophages. We have found that CD36 differently regulates activation of mouse macrophages by S-LPS versus R-LPS. The ability of CD36 to substitute for CD14 in loading R-LPS, but not S-LPS onto TLR4/MD-2 allows CD14-independent macrophage responses to R-LPS. Conversely, S-LPS, but not R-LPS effectively stimulates CD14 binding to CD36, which favors S-LPS transfer from CD14 onto TLR4/MD-2 under conditions of low CD14 occupancy with S-LPS in serum-free medium. In contrast, in the presence of serum, CD36 reduces S-LPS binding to TLR4/MD-2 and the subsequent MyD88-dependent signaling, by mediating internalization of S-LPS/CD14 complexes. Additionally, CD36 positively regulates activation of TRIF-dependent signaling by both S-LPS and R-LPS, by promoting TLR4/MD-2 endocytosis. In contrast, we have found that SR-A does not function as a S-LPS receptor. Thus, by co-operating with CD14 in both R- and S-LPS loading onto TLR4/MD-2, CD36 can enhance the sensitivity of tissue-resident macrophages in detecting infections by Gram-negative bacteria. However, in later phases, following influx of serum to the infection site, the CD36-mediated negative regulation of MyD88-dependent branch of S-LPS-induced TLR4 signaling might constitute a mechanism to prevent an excessive inflammatory response, while preserving the adjuvant effect of S-LPS for adaptive immunity.
Collapse
Affiliation(s)
- Rafał Biedroń
- Department of Immunology, Jagiellonian University Medical College, Kraków, Poland
| | - Angelika Peruń
- Department of Immunology, Jagiellonian University Medical College, Kraków, Poland
| | - Szczepan Józefowski
- Department of Immunology, Jagiellonian University Medical College, Kraków, Poland
- * E-mail:
| |
Collapse
|
21
|
Cauwelaert ND, Desbien AL, Hudson TE, Pine SO, Reed SG, Coler RN, Orr MT. The TLR4 Agonist Vaccine Adjuvant, GLA-SE, Requires Canonical and Atypical Mechanisms of Action for TH1 Induction. PLoS One 2016; 11:e0146372. [PMID: 26731269 PMCID: PMC4701231 DOI: 10.1371/journal.pone.0146372] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
The Toll-like receptor 4 agonist glucopyranosyl lipid adjuvant formulated in a stable emulsion (GLA-SE) promotes strong TH1 and balanced IgG1/IgG2 responses to protein vaccine antigens. This enhanced immunity is sufficient to provide protection against many diseases including tuberculosis and leishmaniasis. To better characterize the adjuvant action it is important to understand how the different cytokines and transcription factors contribute to the initiation of immunity. In the present study using T-bet-/- and IL-12-/- mice and a blocking anti-IFNαR1 monoclonal antibody, we define mechanisms of adjuvant activity of GLA-SE. In accordance with previous studies of TLR4 agonist based adjuvants, we found that TH1 induction via GLA-SE was completely dependent upon T-bet, a key transcription factor for IFNγ production and TH1 differentiation. Consistent with this, deficiency of IL-12, a cytokine canonical to TH1 induction, ablated TH1 induction via GLA-SE. Finally we demonstrate that the innate immune response to GLA-SE, including rapid IFNγ production by memory CD8+ T cells and NK cells, was contingent on type I interferon, a cytokine group whose association with TH1 induction is contextual, and that they contributed to the adjuvant activity of GLA-SE.
Collapse
Affiliation(s)
| | - Anthony L. Desbien
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Thomas E. Hudson
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Samuel O. Pine
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | - Steven G. Reed
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Rhea N. Coler
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Mark T. Orr
- Infectious Disease Research Institute, Seattle, Washington, United States of America
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
22
|
Wu M, Wang H, Shi J, Sun J, Duan Z, Li Y, Li J, Hu N, Wei Y, Chen Y, Hu Y. Gene expression profiles identify both MyD88-independent and MyD88-dependent pathways involved in the maturation of dendritic cells mediated by heparan sulfate: a novel adjuvant. Hum Vaccin Immunother 2015; 10:3711-21. [PMID: 25668674 DOI: 10.4161/21645515.2014.980682] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The traditional vaccine adjuvant research is mainly based on the trial and error method, and the mechanisms underlying the immune system stimulation remaining largely unknown. We previously demonstrated that heparan sulfate (HS), a TLR-4 ligand and endogenous danger signal, effectively enhanced humoral and cellular immune responses in mice immunized by HBsAg. This study aimed to evaluate whether HS induces better humoral immune responses against inactivated Hepatitis A or Rabies Vaccines, respectively, compared with traditional adjuvants (e.g. Alum and complete Freund's adjuvant). In order to investigate the molecular mechanisms of its adjuvanticity, the gene expression pattern of peripheral blood monocytes derived DCs (dendritic cells) stimulated with HS was analyzed at different times points. Total RNA was hybridized to Agilent SurePrint G3 Human Gene Expression 8×60 K one-color oligo-microarray. Through intersection analysis of the microarray results, we found that the Toll-like receptor signaling pathway was significantly activated, and NF-kB, TRAF3 and IRF7 were activated as early as 12 h, and MyD88 was activated at 48 h post-stimulation. Furthermore, the expression of the surface marker CD83 and the co-stimulatory molecules CD80 and CD86 was up-regulated as early as 24 h. Therefore, we speculated that HS-induced human monocyte-derived DC maturation may occur through both MyD88-independent and dependent pathways, but primarily through the former (TRIF pathway). These data provide an important basis for understanding the mechanisms underlying HS enhancement of the immune response.
Collapse
Key Words
- DCs, Dendritic cells
- DEGs, differentially expressed genes
- GO, Gene Ontology
- HAV, hepatitis A virus
- HBsAg, hepatitis B surface antigen
- HS, heparan sulfate
- IRF7, interferon regulatory factor 7
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- MyD88, myeloid differentiation primary response 88
- NF-kB, nuclear factor-kappa B
- Rab/Vac, Rabies Vaccine
- TRAF3, TNF receptor-associated factor 3
- dendritic cells
- gene expression profile
- heparan sulfate
- humoral immune response
- toll-like receptor signaling pathway
- vaccine adjuvant
Collapse
Affiliation(s)
- Meini Wu
- a Institute of Medical Biology; Chinese Academy of Medical Sciences and Peking Union Medical College ; Kunming , China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Bhunia D, Pallavi PMC, Bonam SR, Reddy SA, Verma Y, Halmuthur MSK. Design, Synthesis, and Evaluation of Novel 1,2,3-Triazole-Tethered Glycolipids as Vaccine Adjuvants. Arch Pharm (Weinheim) 2015; 348:689-703. [DOI: 10.1002/ardp.201500143] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/26/2015] [Accepted: 07/27/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Debabrata Bhunia
- Vaccine Immunology Laboratory; Natural Products Chemistry Division; CSIR-Indian Institute of Chemical Technology; Hyderabad India
| | - Preethi M. C. Pallavi
- Vaccine Immunology Laboratory; Natural Products Chemistry Division; CSIR-Indian Institute of Chemical Technology; Hyderabad India
| | - Srinivasa Reddy Bonam
- Vaccine Immunology Laboratory; Natural Products Chemistry Division; CSIR-Indian Institute of Chemical Technology; Hyderabad India
| | - Sandeep A. Reddy
- Vaccine Immunology Laboratory; Natural Products Chemistry Division; CSIR-Indian Institute of Chemical Technology; Hyderabad India
| | - Yogesh Verma
- Vaccine Immunology Laboratory; Natural Products Chemistry Division; CSIR-Indian Institute of Chemical Technology; Hyderabad India
| | - M. Sampath Kumar Halmuthur
- Vaccine Immunology Laboratory; Natural Products Chemistry Division; CSIR-Indian Institute of Chemical Technology; Hyderabad India
| |
Collapse
|
24
|
Zariri A, van der Ley P. Biosynthetically engineered lipopolysaccharide as vaccine adjuvant. Expert Rev Vaccines 2015; 14:861-76. [PMID: 25797360 DOI: 10.1586/14760584.2015.1026808] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Lipopolysaccharide (LPS), a dominant component of the Gram-negative bacterial outer membrane, is a strong activator of the innate immune system, and thereby an important determinant in the adaptive immune response following bacterial infection. This adjuvant activity can be harnessed following immunization with bacteria-derived vaccines that naturally contain LPS, and when LPS or molecules derived from it are added to purified vaccine antigens. However, the downside of the strong biological activity of LPS is its ability to contribute to vaccine reactogenicity. Modification of the LPS structure allows triggering of a proper immune response needed in a vaccine against a particular pathogen while at the same time lowering its toxicity. Extensive modifications to the basic structure are possible by using our current knowledge of bacterial genes involved in LPS biosynthesis and modification. This review focuses on biosynthetic engineering of the structure of LPS and implications of these modifications for generation of safe adjuvants.
Collapse
Affiliation(s)
- Afshin Zariri
- Institute for Translational Vaccinology (InTraVacc), Antonie van Leeuwenhoeklaan 9, 3721 MA Bilthoven, The Netherlands
| | | |
Collapse
|
25
|
Synthetic Toll-like receptor 4 (TLR4) and TLR7 ligands as influenza virus vaccine adjuvants induce rapid, sustained, and broadly protective responses. J Virol 2015; 89:3221-35. [PMID: 25568203 DOI: 10.1128/jvi.03337-14] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Current vaccines against influenza virus infection rely on the induction of neutralizing antibodies targeting the globular head of the viral hemagglutinin (HA). Protection against seasonal antigenic drift or sporadic pandemic outbreaks requires further vaccine development to induce cross-protective humoral responses, potentially to the more conserved HA stalk region. Here, we present a novel viral vaccine adjuvant comprised of two synthetic ligands for Toll-like receptor 4 (TLR4) and TLR7. 1Z105 is a substituted pyrimido[5,4-b]indole specific for the TLR4-MD2 complex, and 1V270 is a phospholipid-conjugated TLR7 agonist. Separately, 1Z105 induces rapid Th2-associated IgG1 responses, and 1V270 potently generates Th1 cellular immunity. 1Z105 and 1V270 in combination with recombinant HA from the A/Puerto Rico/8/1934 strain (rPR/8 HA) effectively induces rapid and sustained humoral immunity that is protective against lethal challenge with a homologous virus. More importantly, immunization with the combined adjuvant and rPR/8 HA, a commercially available split vaccine, or chimeric rHA antigens significantly improves protection against both heterologous and heterosubtypic challenge viruses. Heterosubtypic protection is associated with broadly reactive antibodies to HA stalk epitopes. Histological examination and cytokine profiling reveal that intramuscular (i.m.) administration of 1Z105 and 1V270 is less reactogenic than a squalene-based adjuvant, AddaVax. In summary, the combination of 1Z105 and 1V270 with a recombinant HA induces rapid, long-lasting, and balanced Th1- and Th2-type immunity; demonstrates efficacy in a variety of murine influenza virus vaccine models assaying homologous, heterologous, and heterosubtypic challenge viruses; and has an excellent safety profile. IMPORTANCE Novel adjuvants are needed to enhance immunogenicity and increase the protective breadth of influenza virus vaccines to reduce the seasonal disease burden and ensure pandemic preparedness. We show here that the combination of synthetic Toll-like receptor 4 (TLR4) and TLR7 ligands is a potent adjuvant for recombinant influenza virus hemagglutinin, inducing rapid and sustained immunity that is protective against influenza viruses in homologous, heterologous, and heterosubtypic challenge models. Combining TLR4 and TLR7 ligands balances Th1- and Th2-type immune responses for long-lived cellular and neutralizing humoral immunity against the viral hemagglutinin. The combined adjuvant has an attractive safety profile and the potential to augment seasonal-vaccine breadth, contribute to a broadly neutralizing universal vaccine formulation, and improve response time in an emerging pandemic.
Collapse
|
26
|
Kolb JP, Casella CR, SenGupta S, Chilton PM, Mitchell TC. Type I interferon signaling contributes to the bias that Toll-like receptor 4 exhibits for signaling mediated by the adaptor protein TRIF. Sci Signal 2014; 7:ra108. [PMID: 25389373 PMCID: PMC4459894 DOI: 10.1126/scisignal.2005442] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Signaling by Toll-like receptor 4 (TLR4) is mediated by either of two adaptor proteins: myeloid differentiation marker 88 (MyD88) or Toll-interleukin-1 (IL-1) receptor (TIR) domain-containing adaptor inducing interferon-β (TRIF). Whereas MyD88-mediated signaling leads to proinflammatory responses, TRIF-mediated signaling leads to less toxic immunostimulatory responses that are beneficial in boosting vaccine responses. The hypothesis that monophosphorylated lipid A structures act as TRIF-biased agonists of TLR4 offered a potential mechanism to explain their clinical value as vaccine adjuvants, but studies of TRIF-biased agonists have been contradictory. In experiments with mouse dendritic cells, we found that irrespective of the agonist used, TLR4 functioned as a TRIF-biased signaling system through a mechanism that depended on the autocrine and paracrine effects of type I interferons. The TLR4 agonist synthetic lipid A induced expression of TRIF-dependent genes at lower concentrations than were necessary to induce the expression of genes that depend on MyD88-mediated signaling. Blockade of type I interferon signaling selectively decreased the potency of lipid A (increased the concentration required) in inducing the expression of TRIF-dependent genes, thereby eliminating adaptor bias. These data may explain how high-potency TLR4 agonists can act as clinically useful vaccine adjuvants by selectively activating TRIF-dependent signaling events required for immunostimulation, without or only weakly activating potentially harmful MyD88-dependent inflammatory responses.
Collapse
Affiliation(s)
- Joseph P Kolb
- Department of Microbiology and Immunology, University of Louisville School of Medicine, 570 South Preston Street, Louisville, KY 40202, USA. Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Carolyn R Casella
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Shuvasree SenGupta
- Department of Microbiology and Immunology, University of Louisville School of Medicine, 570 South Preston Street, Louisville, KY 40202, USA. Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Paula M Chilton
- Department of Microbiology and Immunology, University of Louisville School of Medicine, 570 South Preston Street, Louisville, KY 40202, USA. Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Thomas C Mitchell
- Department of Microbiology and Immunology, University of Louisville School of Medicine, 570 South Preston Street, Louisville, KY 40202, USA. Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
27
|
Pouliot K, Buglione-Corbett R, Marty-Roix R, Montminy-Paquette S, West K, Wang S, Lu S, Lien E. Contribution of TLR4 and MyD88 for adjuvant monophosphoryl lipid A (MPLA) activity in a DNA prime-protein boost HIV-1 vaccine. Vaccine 2014; 32:5049-56. [PMID: 25045815 PMCID: PMC10687719 DOI: 10.1016/j.vaccine.2014.07.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 06/17/2014] [Accepted: 07/08/2014] [Indexed: 12/19/2022]
Abstract
Recombinant protein vaccines are commonly formulated with an immune-stimulatory compound, or adjuvant, to boost immune responses to a particular antigen. Recent studies have shown that, through recognition of molecular motifs, receptors of the innate immune system are involved in the functions of adjuvants to generate and direct adaptive immune responses. However, it is not clear to which degree those receptors are also important when the adjuvant is used as part of a novel heterologous prime-boost immunization process in which the priming and boosting components are not the same type of vaccines. In the current study, we compared the immune responses elicited by a pentavalent HIV-1 DNA prime-protein boost vaccine in mice deficient in either Toll-like receptor 4 (TLR4) or myeloid differentiation primary response gene 88 (MyD88) to wildtype mice. HIV gp120 protein administered in the boost phase was formulated with either monophosphoryl lipid A (MPLA), QS-21, or Al(OH)3. Endpoint antibody titer, serum cytokine response and T-cell memory response were assessed. Neither TLR4 nor MyD88 deficiency had a significant effect on the immune response of mice given vaccine formulated with QS-21 or Al(OH)3. However, TLR4- and MyD88-deficiency decreased both the antibody and T-cell responses in mice administered HIV gp120 formulated with MPLA. These results further our understanding of the activation of TLR4 and MyD88 by MPLA in the context of a DNA prime/protein boost immunization strategy.
Collapse
Affiliation(s)
- Kimberly Pouliot
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Rachel Buglione-Corbett
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Robyn Marty-Roix
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Sara Montminy-Paquette
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States
| | - Kim West
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shixia Wang
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Shan Lu
- Laboratory of Nucleic Acid Vaccines, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Egil Lien
- Division of Infectious Diseases and Immunology, Program in Innate Immunity, Worcester, MA 01605, United States; Centre of Molecular Inflammation Research, Dept. of Cancer and Molecular Medicine, NTNU, 7491 Trondheim, Norway.
| |
Collapse
|
28
|
Bohannon JK, Hernandez A, Enkhbaatar P, Adams WL, Sherwood ER. The immunobiology of toll-like receptor 4 agonists: from endotoxin tolerance to immunoadjuvants. Shock 2014; 40:451-62. [PMID: 23989337 DOI: 10.1097/shk.0000000000000042] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lipopolysaccharide (LPS, endotoxin) is a structural component of the gram-negative outer membrane. The lipid A moiety of LPS binds to the LPS receptor complex expressed by leukocytes, endothelial cells, and parenchymal cells and is the primary component of gram-negative bacteria that is recognized by the immune system. Activation of the LPS receptor complex by native lipid A induces robust cytokine production, leukocyte activation, and inflammation, which is beneficial for clearing bacterial infections at the local level but can cause severe systemic inflammation and shock at higher challenge doses. Interestingly, prior exposure to LPS renders the host resistant to shock caused by subsequent LPS challenge, a phenomenon known as endotoxin tolerance. Treatment with lipid A has also been shown to augment the host response to infection and to serve as a potent vaccine adjuvant. However, the adverse effects associated with the pronounced inflammatory response limit the use of native lipid A as a clinical immunomodulator. More recently, analogs of lipid A have been developed that possess attenuated proinflammatory activity but retain attractive immunomodulatory properties. The lipid A analog monophosphoryl lipid A exhibits approximately 1/1,000th of the toxicity of native lipid A but retains potent immunoadjuvant activity. As such, monophosphoryl lipid A is currently used as an adjuvant in several human vaccine preparations. Because of the potency of lipid A analogs as immunoadjuvants, numerous laboratories are actively working to identify and develop new lipid A mimetics and to optimize their efficacy and safety. Based on those characteristics, lipid A analogs represent an attractive family of immunomodulators.
Collapse
Affiliation(s)
- Julia K Bohannon
- *Department of Anesthesiology, Vanderbilt University Medical Center, Nashville, Tennessee; †Department of Anesthesiology, The University of Texas Medical Branch, Galveston, Texas; and ‡School of Medicine, The University of Tennessee Health Science Center, Memphis; and §Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | | | | |
Collapse
|
29
|
Bergmann-Leitner ES, Leitner WW. Adjuvants in the Driver's Seat: How Magnitude, Type, Fine Specificity and Longevity of Immune Responses Are Driven by Distinct Classes of Immune Potentiators. Vaccines (Basel) 2014; 2:252-96. [PMID: 26344620 PMCID: PMC4494256 DOI: 10.3390/vaccines2020252] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/20/2014] [Accepted: 03/28/2014] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which vaccine adjuvants enhance immune responses has historically been considered to be the creation of an antigen depot. From here, the antigen is slowly released and provided to immune cells over an extended period of time. This "depot" was formed by associating the antigen with substances able to persist at the injection site, such as aluminum salts or emulsions. The identification of Pathogen-Associated Molecular Patterns (PAMPs) has greatly advanced our understanding of how adjuvants work beyond the simple concept of extended antigen release and has accelerated the development of novel adjuvants. This review focuses on the mode of action of different adjuvant classes in regards to the stimulation of specific immune cell subsets, the biasing of immune responses towards cellular or humoral immune response, the ability to mediate epitope spreading and the induction of persistent immunological memory. A better understanding of how particular adjuvants mediate their biological effects will eventually allow them to be selected for specific vaccines in a targeted and rational manner.
Collapse
Affiliation(s)
- Elke S Bergmann-Leitner
- US Military Malaria Research Program, Malaria Vaccine Branch, 503 Robert Grant Ave, 3W65, Silver Spring, MD 20910, USA.
| | - Wolfgang W Leitner
- Division on Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 6610 Rockledge Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Calil IL, Zarpelon AC, Guerrero ATG, Alves-Filho JC, Ferreira SH, Cunha FQ, Cunha TM, Verri WA. Lipopolysaccharide induces inflammatory hyperalgesia triggering a TLR4/MyD88-dependent cytokine cascade in the mice paw. PLoS One 2014; 9:e90013. [PMID: 24595131 PMCID: PMC3940714 DOI: 10.1371/journal.pone.0090013] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Accepted: 01/29/2014] [Indexed: 01/18/2023] Open
Abstract
Inflammatory pain can be triggered by different stimuli, such as trauma, radiation, antigen and infection. In a model of inflammatory pain caused by infection, injection in the mice paw of lipopolysaccharide (LPS), a Toll-like receptor 4 (TLR4) agonist, produces mechanical hyperalgesia. We identify here the TLR4 linked signaling pathways that elicit this response. Firstly, LPS paw injection in wild type (WT) mice produced mechanical hyperalgesia that was not altered in TRIF-/- mice. On the other hand, this response was absent in TLR4 mutant and MyD88 null mice and reduced in TNFR1 null mice. Either an IL-1 receptor antagonist, anti-KC/CXCL1 antibody, indomethacin or guanethidine injection also lessened this response. Moreover, LPS-induced time dependent increases in TNF-α, KC/CXCL1 and IL-1β expression in the mice paw, which were absent in TLR4 mutant and MyD88 null mice. Furthermore, in TNFR1 deficient mice, the LPS-induced rises in KC/CXCL1 and IL-1β release were less than in their wild type counterpart. LPS also induced increase of myeloperoxidase activity in the paw skin, which was inhibited in TLR4 mutant and MyD88 null mice, and not altered in TRIF-/- mice. These results suggest that LPS-induced inflammatory pain in mice is solely dependent on the TLR4/MyD88 rather than the TLR4/TRIF signaling pathway. This pathway triggers pronociceptive cytokine TNF-α release that in turn mediates rises in KC/CXCL1 and IL-1β expression. Finally, these cytokines might be involved in stimulating production of directly-acting hyperalgesic mediators such as prostaglandins and sympathomimetic amine.
Collapse
Affiliation(s)
- Igor L. Calil
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ana C. Zarpelon
- Departamento de Patologia, Centro de Ciências Biologicas, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
| | | | - Jose C. Alves-Filho
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Sergio H. Ferreira
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q. Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thiago M. Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- * E-mail: (WAV); (TMC)
| | - Waldiceu A. Verri
- Departamento de Patologia, Centro de Ciências Biologicas, Universidade Estadual de Londrina, Londrina, Paraná, Brazil
- * E-mail: (WAV); (TMC)
| |
Collapse
|
31
|
Han JE, Wui SR, Kim KS, Cho YJ, Cho WJ, Lee NG. Characterization of the structure and immunostimulatory activity of a vaccine adjuvant, de-O-acylated lipooligosaccharide. PLoS One 2014; 9:e85838. [PMID: 24465739 PMCID: PMC3899070 DOI: 10.1371/journal.pone.0085838] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 12/02/2013] [Indexed: 12/20/2022] Open
Abstract
Lipopolysaccharide (LPS) is a major component of the outer membrane of Gram-negative bacteria. LPS elicits strong immunopathological responses during bacterial infection, and the lipid A moiety of LPS is responsible for this immunostimulatory activity. Lipid A exerts its biological activity by sending signals via TLR4 present on immune cells, and TLR4 agonists have been a target for vaccine adjuvant. Previously, we demonstrated an adjuvant activity of deacylated lipooligosaccharide (dLOS) to viral and bacterial antigens. In this study, we characterized the chemical structure of dLOS and evaluated its immunostimulatory activity on mouse and human immune cells in comparison with monophosphoryl lipid A (MPL). dLOS consists of a core oligosaccharide lacking the terminal glucose residue, a glucosamine disaccharide with two phosphate groups, and two N-linked acyl groups. dLOS was similar to MPL in induction of cytokine production in mouse peritoneal macrophages, but was a more potent activator in human monocytes and dendritic cells (DCs). Results of an analysis of allogeneic T cell responses revealed that dLOS induces Th1, Th2, and Th17-type immune responses in a dose-dependent manner. The immunostimulatory activities of dLOS were completely abrogated in TLR4−/− mice, which confirms its TLR4-dependency. These results suggest that in the presence of the core oligosaccharide, O-linked acyl groups of LPS are dispensable for activating the TLR4 signaling pathway. dLOS did not cause any pathological effects or death at 0.25, 0.5, or 1 mg per kg body weight in mice in the acute toxicity tests. This result suggests that dLOS has a low toxicity. dLOS should be considered for further development as a safe and effective adjuvant for human vaccines.
Collapse
MESH Headings
- Acylation
- Adjuvants, Immunologic/chemistry
- Adjuvants, Immunologic/pharmacology
- Animals
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Cytokines/blood
- Cytokines/immunology
- Cytokines/metabolism
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dose-Response Relationship, Drug
- Female
- Flow Cytometry
- Humans
- Immunoblotting
- Lipopolysaccharides/chemistry
- Lipopolysaccharides/immunology
- Lipopolysaccharides/pharmacology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Molecular Structure
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/metabolism
- Toll-Like Receptor 4/deficiency
- Toll-Like Receptor 4/genetics
- Toll-Like Receptor 4/immunology
- Vaccines/immunology
Collapse
Affiliation(s)
- Ji Eun Han
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
| | - Seo Ri Wui
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
| | - Kwang Sung Kim
- Research & Development Center, EyeGene, Seoul, Republic of Korea
| | - Yang Je Cho
- Research & Development Center, EyeGene, Seoul, Republic of Korea
| | - Wan Je Cho
- Yonsei University Gangnam Severance Hospital, Seoul, Republic of Korea
| | - Na Gyong Lee
- Department of Bioscience & Biotechnology, College of Bioscience, Sejong University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
32
|
Adjuvant activity of naturally occurring monophosphoryl lipopolysaccharide preparations from mucosa-associated bacteria. Infect Immun 2013; 81:3317-25. [PMID: 23798540 DOI: 10.1128/iai.01150-12] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Natural heterogeneity in the structure of the lipid A portion of lipopolysaccharide (LPS) produces differential effects on the innate immune response. Gram-negative bacterial species produce LPS structures that differ from the classic endotoxic LPS structures. These differences include hypoacylation and hypophosphorylation of the diglucosamine backbone, both differences known to decrease LPS toxicity. The effect of decreased toxicity on the adjuvant properties of many of these LPS structures has not been fully explored. Here we demonstrate that two naturally produced forms of monophosphorylated LPS, from the mucosa-associated bacteria Bacteroides thetaiotaomicron and Prevotella intermedia, function as immunological adjuvants for antigen-specific immune responses. Each form of mucosal LPS increased vaccination-initiated antigen-specific antibody titers in both quantity and quality when given simultaneously with vaccine antigen preparations. Interestingly, adjuvant effects on initial T cell clonal expansion were selective for CD4 T cells. No significant increase in CD8 T cell expansion was detected. MyD88/Toll-like receptor 4 (TLR4) and TRIF/TLR4 signaling pathways showed equally decreased signaling with the LPS forms studied here as with endotoxic LPS or detoxified monophosphorylated lipid A (MPLA). Natural monophosphorylated LPS from mucosa-associated bacteria functions as a weak but effective adjuvant for specific immune responses, with preferential effects on antibody and CD4 T cell responses over CD8 T cell responses.
Collapse
|