1
|
Pizzuto M, Hurtado-Navarro L, Molina-Lopez C, Soubhye J, Gelbcke M, Rodriguez-Lopez S, Ruysschaert JM, Schroder K, Pelegrin P. Ornithine lipid is a partial TLR4 agonist and NLRP3 activator. Cell Rep 2024; 43:114788. [PMID: 39340778 DOI: 10.1016/j.celrep.2024.114788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/13/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Gram-negative bacterial lipopolysaccharides (LPSs) trigger inflammatory reactions through Toll-like receptor 4 (TLR4) and prime myeloid cells for inflammasome activation. In phosphate-limited environments, bacteria reduce LPS and other phospholipid production and synthesize phosphorus-free alternatives such as amino-acid-containing lipids like the ornithine lipid (OL). This adaptive strategy conserves phosphate for other essential cellular processes and enhances bacterial survival in host environments. While OL is implicated in bacterial pathogenicity, the mechanism is unclear. Using primary murine macrophages and human mononuclear cells, we elucidate that OL activates TLR4 and induces potassium efflux-dependent nucleotide-binding domain and leucine-rich repeat-containing pyrin protein 3 (NLRP3) activation. OL upregulates the expression of NLRP3 and pro-interleukin (IL)-1β and induces cytokine secretion in primed and unprimed cells. By contrast, in the presence of LPS, OL functions as a partial TLR4 antagonist and reduces LPS-induced cytokine secretion. We thus suggest that in phosphate-depleted environments, OL replaces LPS bacterial immunogenicity, while constitutively present OL may allow bacteria to escape immune surveillance.
Collapse
Affiliation(s)
- Malvina Pizzuto
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; Structure and Function of Biological Membranes Laboratory, Université Libre de Bruxelles, 1050 Brussels, Belgium; Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4067, Australia.
| | - Laura Hurtado-Navarro
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Cristina Molina-Lopez
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Jalal Soubhye
- Department of Pharmacognosy, Bioanalysis and Drug Discovery, Faculty of Pharmacy, Université Libre De Bruxelles, 1050 Brussels, Belgium
| | - Michel Gelbcke
- Department of Pharmacognosy, Bioanalysis and Drug Discovery, Faculty of Pharmacy, Université Libre De Bruxelles, 1050 Brussels, Belgium
| | - Silvia Rodriguez-Lopez
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain
| | - Jean-Marie Ruysschaert
- Structure and Function of Biological Membranes Laboratory, Université Libre de Bruxelles, 1050 Brussels, Belgium
| | - Kate Schroder
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4067, Australia
| | - Pablo Pelegrin
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), 30120 Murcia, Spain; Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, 30120 Murcia, Spain
| |
Collapse
|
2
|
Shah MM, Layhadi JA, Hourcade DE, Fulton WT, Tan TJ, Dunham D, Chang I, Vel MS, Fernandes A, Lee AS, Liu J, Arunachalam PS, Galli SJ, Boyd SD, Pulendran B, Davis MM, O’Hara R, Park H, Mitchell LM, Akk A, Patterson A, Jerath MR, Monroy JM, Ren Z, Kendall PL, Durham SR, Fedina A, Gibbs BF, Agache I, Chinthrajah S, Sindher SB, Heider A, Akdis CA, Shamji MH, Pham CT, Nadeau KC. Elucidating allergic reaction mechanisms in response to SARS-CoV-2 mRNA vaccination in adults. Allergy 2024; 79:2502-2523. [PMID: 39033312 PMCID: PMC11368657 DOI: 10.1111/all.16231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND During the COVID-19 pandemic, novel nanoparticle-based mRNA vaccines were developed. A small number of individuals developed allergic reactions to these vaccines although the mechanisms remain undefined. METHODS To understand COVID-19 vaccine-mediated allergic reactions, we enrolled 19 participants who developed allergic events within 2 h of vaccination and 13 controls, nonreactors. Using standard hemolysis assays, we demonstrated that sera from allergic participants induced stronger complement activation compared to nonallergic subjects following ex vivo vaccine exposure. RESULTS Vaccine-mediated complement activation correlated with anti-polyethelyne glycol (PEG) IgG (but not IgM) levels while anti-PEG IgE was undetectable in all subjects. Depletion of total IgG suppressed complement activation in select individuals. To investigate the effects of vaccine excipients on basophil function, we employed a validated indirect basophil activation test that stratified the allergic populations into high and low responders. Complement C3a and C5a receptor blockade in this system suppressed basophil response, providing strong evidence for complement involvement in vaccine-mediated basophil activation. Single-cell multiome analysis revealed differential expression of genes encoding the cytokine response and Toll-like receptor (TLR) pathways within the monocyte compartment. Differential chromatin accessibility for IL-13 and IL-1B genes was found in allergic and nonallergic participants, suggesting that in vivo, epigenetic modulation of mononuclear phagocyte immunophenotypes determines their subsequent functional responsiveness, contributing to the overall physiologic manifestation of vaccine reactions. CONCLUSION These findings provide insights into the mechanisms underlying allergic reactions to COVID-19 mRNA vaccines, which may be used for future vaccine strategies in individuals with prior history of allergies or reactions and reduce vaccine hesitancy.
Collapse
Affiliation(s)
- Mihir M. Shah
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- These authors contributed equally to this work
| | - Janice A. Layhadi
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
- These authors contributed equally to this work
| | - Dennis E. Hourcade
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
- These authors contributed equally to this work
| | - William T. Fulton
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Tiak Ju Tan
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Diane Dunham
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Iris Chang
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Monica S. Vel
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Andrea Fernandes
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - Alexandra S. Lee
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
| | - James Liu
- Stanford Health Library; Stanford, CA, USA
| | - Prabhu S. Arunachalam
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
| | - Stephen J. Galli
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Scott D. Boyd
- Sean N. Parker Center for Allergy & Asthma Research;
Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
- Department of Pathology, Stanford University School of
Medicine; Stanford, California, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection,
Stanford University; Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford
University School of Medicine; Stanford, California, USA
| | - Ruth O’Hara
- Department of Veteran’s Administration and
Dean’s Office, Stanford University; Stanford, CA, USA
| | - Helen Park
- Veterans Affairs Palo Alto Health Care System; Palo Alto,
CA, USA
| | - Lynne M. Mitchell
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
| | - Antonina Akk
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
| | - Alexander Patterson
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Maya R. Jerath
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jennifer M. Monroy
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Zhen Ren
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Peggy L. Kendall
- Department of Medicine, Division of Allergy and
Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Stephen R. Durham
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Aleksandra Fedina
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
| | - Bernhard F Gibbs
- Department of Human Medicine, School of Medicine and
Health Sciences, Carl von Ossietzky University of Oldenburg; Oldenburg,
Germany
- Canterbury Christ Church University, UK
| | - Ioana Agache
- Faculty of Medicine, Transilvania University; Brasov,
Romania
| | | | | | - Anja Heider
- Swiss Institute of Allergy and Asthma Research (SIAF),
University of Zurich; Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF),
University of Zurich; Davos, Switzerland
| | - Mohamed H. Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical
Immunology, Department of National Heart and Lung Institute, Imperial College
London; London, United Kingdom
- These authors contributed equally to this work
| | - Christine T.N. Pham
- Department of Medicine, Division of Rheumatology,
Washington University School of Medicine; St. Louis, MO, USA
- These authors contributed equally to this work
| | - Kari C. Nadeau
- Harvard T.H. Chan School of Public Health, Harvard
University; Cambridge, Massachusetts
- These authors contributed equally to this work
| |
Collapse
|
3
|
Kawai A, Noda M, Hirata H, Munakata L, Matsuda T, Omata D, Takemura N, Onoe S, Hirose M, Kato T, Saitoh T, Hirai T, Suzuki R, Yoshioka Y. Lipid Nanoparticle with 1,2-Di-O-octadecenyl-3-trimethylammonium-propane as a Component Lipid Confers Potent Responses of Th1 Cells and Antibody against Vaccine Antigen. ACS NANO 2024; 18:16589-16609. [PMID: 38885198 PMCID: PMC11223497 DOI: 10.1021/acsnano.4c00278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/20/2024]
Abstract
Adjuvants are effective tools to enhance vaccine efficacy and control the type of immune responses such as antibody and T helper 1 (Th1)- or Th2-type responses. Several studies suggest that interferon (IFN)-γ-producing Th1 cells play a significant role against infections caused by intracellular bacteria and viruses; however, only a few adjuvants can induce a strong Th1-type immune response. Recently, several studies have shown that lipid nanoparticles (LNPs) can be used as vaccine adjuvants and that each LNP has a different adjuvant activity. In this study, we screened LNPs to develop an adjuvant that can induce Th1 cells and antibodies using a conventional influenza split vaccine (SV) as an antigen in mice. We observed that LNP with 1,2-di-O-octadecenyl-3-trimethylammonium-propane (DOTMA) as a component lipid (DOTMA-LNP) elicited robust SV-specific IgG1 and IgG2 responses compared with SV alone in mice and was as efficient as SV adjuvanted with other adjuvants in mice. Furthermore, DOTMA-LNPs induced robust IFN-γ-producing Th1 cells without inflammatory responses compared to those of other adjuvants, which conferred strong cross-protection in mice. We also demonstrated the high versatility of DOTMA-LNP as a Th1 cell-inducing vaccine adjuvant using vaccine antigens derived from severe acute respiratory syndrome coronavirus 2 and Streptococcus pneumoniae. Our findings suggest the potential of DOTMA-LNP as a safe and effective Th1 cell-inducing adjuvant and show that LNP formulations are potentially potent adjuvants to enhance the effectiveness of other subunit vaccines.
Collapse
Affiliation(s)
- Atsushi Kawai
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masahiro Noda
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Haruki Hirata
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Lisa Munakata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Teppei Matsuda
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Daiki Omata
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Naoki Takemura
- Laboratory
of Bioresponse Regulation, Graduate School
of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Sakura Onoe
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Mika Hirose
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takayuki Kato
- Institute
for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tatsuya Saitoh
- Laboratory
of Bioresponse Regulation, Graduate School
of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Infectious Disease Education and Research, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Global
Center for Medical Engineering and Informatics, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshiro Hirai
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ryo Suzuki
- Laboratory
of Drug and Gene Delivery Research, Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Yasuo Yoshioka
- Laboratory
of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Institute for Open and Transdisciplinary Research
Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center
for Infectious Disease Education and Research, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Global
Center for Medical Engineering and Informatics, Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
- Vaccine
Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, The Research Foundation for Microbial Diseases of
Osaka University, 3-1
Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
4
|
Bayram B, Senarslan DA, Sengel A, Ozturk T, Onur E, Iskesen I. Does remote ischemic preconditioning affect the systemic inflammatory response by modulating presepsin levels? Int J Artif Organs 2024; 47:388-393. [PMID: 38761055 DOI: 10.1177/03913988241255495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
OBJECTIVE We investigated the effect of Remote Ischemic Preconditioning (RIPC) on the inflammatory response during CPB by means of serum presepsin levels at preoperative and postoperative 1st and 24th h. METHODS In this prospective, randomized, cross-sectional study we included 81 patients undergoing coronary artery bypass graft surgery with cardiopulmonary bypass (CPB). Patients were randomized and RIPC was applied to 40 patients in the study group before anesthesia. The remaining 41 patients were determined as the control group. The relationships between RIPC and factors such as presepsin, C-reactive protein (CRP), and leukocyte levels were investigated. RESULTS There was no significant difference between the groups in postoperative leukocyte and CRP values (p = 0.52, p = 0.13, respectively). When the preoperative and postoperative first hour presepsin values of the patients were compared, no significant difference was found in the control group (p = 0.17), but a significant difference was found in the study group (p < 0.05). When the presepsin values were compared between the groups, a significant difference was found only in the postoperative first hour value (p < 0.05). CONCLUSIONS It was observed that RIPC application caused to increase the presepsin levels in the postoperative first hour significantly in the study group (p < 0.05).
Collapse
Affiliation(s)
- Barıs Bayram
- Department of Cardiovascular Surgery, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Turkey
| | | | - Arife Sengel
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Turkey
| | - Tulun Ozturk
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Turkey
| | - Ece Onur
- Department of Medical Biochemistry, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Turkey
| | - Ihsan Iskesen
- Department of Cardiovascular Surgery, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
5
|
Lee J, Woodruff MC, Kim EH, Nam JH. Knife's edge: Balancing immunogenicity and reactogenicity in mRNA vaccines. Exp Mol Med 2023; 55:1305-1313. [PMID: 37430088 PMCID: PMC10394010 DOI: 10.1038/s12276-023-00999-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/26/2023] [Accepted: 03/27/2023] [Indexed: 07/12/2023] Open
Abstract
Since the discovery of messenger RNA (mRNA), there have been tremendous efforts to wield them in the development of therapeutics and vaccines. During the COVID-19 pandemic, two mRNA vaccines were developed and approved in record-breaking time, revolutionizing the vaccine development landscape. Although first-generation COVID-19 mRNA vaccines have demonstrated over 90% efficacy, alongside strong immunogenicity in humoral and cell-mediated immune responses, their durability has lagged compared to long-lived vaccines, such as the yellow fever vaccine. Although worldwide vaccination campaigns have saved lives estimated in the tens of millions, side effects, ranging from mild reactogenicity to rare severe diseases, have been reported. This review provides an overview and mechanistic insights into immune responses and adverse effects documented primarily for COVID-19 mRNA vaccines. Furthermore, we discuss the perspectives of this promising vaccine platform and the challenges in balancing immunogenicity and adverse effects.
Collapse
Affiliation(s)
- Jisun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
- Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| | - Eui Ho Kim
- Viral Immunology Laboratory, Institut Pasteur Korea, Seongnam, 13488, Republic of Korea.
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea.
- BK Plus Department of Biotechnology, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea.
| |
Collapse
|
6
|
TLR4 and MD2 variation among horses with differential TNFα baseline concentrations and response to intravenous lipopolysaccharide infusion. Sci Rep 2023; 13:1486. [PMID: 36707633 PMCID: PMC9883502 DOI: 10.1038/s41598-023-27956-y] [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: 09/15/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
Gram-negative bacterial septicemia is mediated through binding of lipopolysaccharide (LPS) to mammalian toll-like receptor protein 4 (TLR4). TLR4 and its cognate protein, myeloid differentiation factor 2 (MD2) form a heterodimeric complex after binding LPS. This complex induces a cascade of reactions that results in increased proinflammatory cytokine gene expression, including TNFα, which leads to activation of innate immunity. In horses, the immune response to LPS varies widely. To determine if this variation is due to differences in TLR4 or MD2, DNA from 15 healthy adult horses with different TNFα dynamics after experimental intravenous LPS infusion was sequenced across exons of TLR4 and MD2. Haplotypes were constructed for both genes using all identified variants. Four haplotypes were observed for each gene. No significant associations were found between either TNFα baseline concentrations or response to LPS and haplotype; however, there was a significant association (P value = 0.0460) between the baseline TNFα concentration and one MD2 missense variant. Three-dimensional structures of the equine TLR4-MD2-LPS complex were built according to haplotype combinations observed in the study horses, and the implications of missense variants on LPS binding were modeled. Although the sample size was small, there was no evidence that variation in TLR4 or MD2 explains the variability in TNFα response observed after LPS exposure in horses.
Collapse
|
7
|
Da Silva Sanchez AJ, Zhao K, Huayamares SG, Hatit MZC, Lokugamage MP, Loughrey D, Dobrowolski C, Wang S, Kim H, Paunovska K, Kuzminich Y, Dahlman JE. Substituting racemic ionizable lipids with stereopure ionizable lipids can increase mRNA delivery. J Control Release 2023; 353:270-277. [PMID: 36423872 PMCID: PMC9897220 DOI: 10.1016/j.jconrel.2022.11.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 12/03/2022]
Abstract
Lipid nanoparticles (LNPs) have delivered siRNA and mRNA drugs in humans, underscoring the potential impact of improving the therapeutic window of next-generation LNPs. To increase the LNP therapeutic window, we applied lessons from small-molecule chemistry to ionizable lipid design. Specifically, given that stereochemistry often influences small-molecule safety and pharmacokinetics, we hypothesized that the stereochemistry of lipids within an LNP would influence mRNA delivery. We tested this hypothesis in vivo using 128 novel LNPs that included stereopure derivatives of C12-200, an ionizable lipid that when formulated into LNPs delivers RNA in mice and non-human primates but is not used clinically due to its poor tolerability. We found that a novel C12-200-S LNP delivered up to 2.8-fold and 6.1-fold more mRNA in vivo than its racemic and C12-200-R controls, respectively. To identify the potential causes leading to increased delivery, we quantified LNP biophysical traits and concluded that these did not change with stereochemistry. Instead, we found that stereopure LNPs were better tolerated than racemic LNPs in vivo. These data suggest that LNP-mediated mRNA delivery can be improved by designing LNPs to include stereopure ionizable lipids.
Collapse
Affiliation(s)
- Alejandro J Da Silva Sanchez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA; Department of Chemical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Kun Zhao
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sebastian G Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Marine Z C Hatit
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Curtis Dobrowolski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Shuaishuai Wang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Hyejin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yanina Kuzminich
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| |
Collapse
|
8
|
Liang H, Lykins WR, Seydoux E, Guderian JA, Phan T, Fox CB, Orr MT. Formulated Phospholipids as Non-Canonical TLR4 Agonists. Pharmaceutics 2022; 14:2557. [PMID: 36559051 PMCID: PMC9788208 DOI: 10.3390/pharmaceutics14122557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022] Open
Abstract
Immunogenic agents known as adjuvants play a critical role in many vaccine formulations. Adjuvants often signal through Toll-like receptor (TLR) pathways, including formulations in licensed vaccines that target TLR4. While TLR4 is predominantly known for responding to lipopolysaccharide (LPS), a component of Gram-negative bacterial membranes, it has been shown to be a receptor for a number of molecular structures, including phospholipids. Therefore, phospholipid-based pharmaceutical formulations might have off-target effects by signaling through TLR4, confounding interpretation of pharmaceutical bioactivity. In this study we examined the individual components of a clinical stage oil-in-water vaccine adjuvant emulsion (referred to as a stable emulsion or SE) and their ability to signal through murine and human TLR4s. We found that the phospholipid 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) activated TLR4 and elicited many of the same immune phenotypes as canonical TLR4 agonists. This pathway was dependent on the saturation, size, and headgroup of the phospholipid. Interestingly, DMPC effects on human cells were evident but overall appeared less impactful than emulsion oil composition. Considering the prevalence of DMPC and other phospholipids used across the pharmaceutical space, these findings may contextualize off-target innate immune responses that could impact preclinical and clinical development.
Collapse
Affiliation(s)
- Hong Liang
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - William R. Lykins
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Emilie Seydoux
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Jeffrey A. Guderian
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Tony Phan
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
| | - Christopher B. Fox
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA 98195, USA
| | - Mark T. Orr
- Access to Advanced Health Institute (AAHI), 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102, USA
- Department of Global Health, University of Washington, 3980 15th Ave NE, Seattle, WA 98195, USA
| |
Collapse
|
9
|
Scherm MJ, Gangloff M, Gay NJ. Activation of Toll-like receptor 4 by Ebola virus-shed glycoprotein is direct and requires the internal fusion loop but not glycosylation. Cell Rep 2022; 41:111562. [PMID: 36288690 PMCID: PMC9637988 DOI: 10.1016/j.celrep.2022.111562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 09/16/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
Infection by the Ebola virus, a member of the Filoviridae family of RNA viruses, leads to acute viral hemorrhagic fever. End-stage Ebola virus disease is characterized by a cytokine storm that causes tissue damage, vascular disintegration, and multi-organ failure. Previous studies showed that a shed form of the viral spike glycoprotein (sGP1,2) drives this hyperinflammatory response by activating Toll-like receptor 4 (TLR4). Here, we find that glycosylation is not required for activation of TLR4 by sGP1,2 and identify the internal fusion loop (IFL) as essential for inflammatory signaling. sGP1,2 competes with lipid antagonists of TLR4, and the IFL interacts directly with TLR4 and co-receptor MD2. Together, these findings indicate that sGP1,2 activates TLR4 analogously to bacterial agonist lipopolysaccharide (LPS) by binding into a hydrophobic pocket in MD2 and promoting the formation of an active heterotetramer. This conclusion is supported by docking studies that predict binding sites for sGP1,2 on TLR4 and MD2.
Collapse
Affiliation(s)
- Michael J. Scherm
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Monique Gangloff
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK
| | - Nicholas J. Gay
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1GA, UK,Corresponding author
| |
Collapse
|
10
|
Pizzuto M, Pelegrin P, Ruysschaert JM. Lipid-protein interactions regulating the canonical and the non-canonical NLRP3 inflammasome. Prog Lipid Res 2022; 87:101182. [PMID: 35901922 DOI: 10.1016/j.plipres.2022.101182] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/25/2022] [Accepted: 07/24/2022] [Indexed: 01/05/2023]
Abstract
The inflammatory response is a complex regulated effector mechanism of the innate immune system that is initiated after tissue injury or infection. The NLRP3 inflammasome is an important initiator of inflammation by regulating the activation of caspase-1, the maturation of pro-inflammatory cytokines and the induction of pyroptotic cell death. Numerous studies demonstrate that the NLRP3 inflammasome could be modulated by lipids, existing a relation between lipids and the activation of different inflammatory processes. In this review we will summarize how the mechanism of NLRP3 inflammasome activation is regulated by different lipids and how these lipids control specific cellular localization of NLRP3 during activation. Although being a cytosolic protein, NLRP3 interacts with lipids accessible in neighbor membranes. Also, the modulation of NLRP3 by endogenous lipids has been found causative of different metabolic diseases and bacterial-pathogenic lipids lead to NLRP3 activation during infection. The understanding of the modulation of the NLRP3 inflammasome by lipids has resulted not only in a better knowledge about the mechanism of NLRP3 activation and its implication in disease, but also opens a new avenue for the development of novel therapeutics and vaccines, as NLRP3 could be modulated by synthetic lipids used as adjuvants.
Collapse
Affiliation(s)
- Malvina Pizzuto
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain; Laboratoire de Structure et Fonction des Membranes Biologiques, Université Libre de Bruxelles, Brussels, Belgium.
| | - Pablo Pelegrin
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain; Department of Biochemistry and Molecular Biology B and Immunology, Faculty of Biology, University of Murcia, Spain.
| | - Jean-Marie Ruysschaert
- Laboratoire de Structure et Fonction des Membranes Biologiques, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
11
|
Won T, Gilotra NA, Wood MK, Hughes DM, Talor MV, Lovell J, Milstone AM, Steenbergen C, Čiháková D. Increased Interleukin 18-Dependent Immune Responses Are Associated With Myopericarditis After COVID-19 mRNA Vaccination. Front Immunol 2022; 13:851620. [PMID: 35251049 PMCID: PMC8894592 DOI: 10.3389/fimmu.2022.851620] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/31/2022] [Indexed: 12/25/2022] Open
Abstract
Myocarditis and myopericarditis may occur after COVID-19 vaccination with an incidence of two to twenty cases per 100,000 individuals, but underlying mechanisms related to disease onset and progression remain unclear. Here, we report a case of myopericarditis following the first dose of the mRNA-1273 COVID-19 vaccine in a young man who had a history of mild COVID-19 three months before vaccination. The patient presented with chest pain, elevated troponin I level, and electrocardiogram abnormality. His endomyocardial biopsy revealed diffuse CD68+ cell infiltration. We characterized the immune profile of the patient using multiplex cytokine assay and flow cytometry analysis. Sex-matched vaccinated individuals and healthy individuals were used as controls. IL-18 and IL-27, Th1-type cytokines, were highly increased in the patient with COVID-19 vaccine-related myopericarditis compared with vaccinated controls who experienced no cardiac complications. In the patient, circulating NK cells and T cells showed an activated phenotype and mRNA profile, and monocytes expressed increased levels of IL-18 and its upstream NLRP3 inflammasome. We found that recombinant IL-18 administration into mice caused mild cardiac dysfunction and activation of NK cells and T cells in the hearts, similar to the findings in the patient with myopericarditis after COVID-19 mRNA vaccination. Collectively, myopericarditis following COVID-19 mRNA vaccination may be associated with increased IL-18-mediated immune responses and cardiotoxicity.
Collapse
Affiliation(s)
- Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nisha Aggarwal Gilotra
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Megan Kay Wood
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - David Matthew Hughes
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, United States
| | - Monica Vladut Talor
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jana Lovell
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Aaron Michael Milstone
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Charles Steenbergen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
- *Correspondence: Daniela Čiháková, ; orcid.org/0000-0002-8713-2860
| |
Collapse
|
12
|
Piccioni A, Santoro MC, de Cunzo T, Tullo G, Cicchinelli S, Saviano A, Valletta F, Pascale MM, Candelli M, Covino M, Franceschi F. Presepsin as Early Marker of Sepsis in Emergency Department: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57080770. [PMID: 34440976 PMCID: PMC8398764 DOI: 10.3390/medicina57080770] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 02/05/2023]
Abstract
The diagnosis and treatment of sepsis have always been a challenge for the physician, especially in critical care setting such as emergency department (ED), and currently sepsis remains one of the major causes of mortality. Although the traditional definition of sepsis based on systemic inflammatory response syndrome (SIRS) criteria changed in 2016, replaced by the new criteria of SEPSIS-3 based on organ failure evaluation, early identification and consequent early appropriated therapy remain the primary goal of sepsis treatment. Unfortunately, currently there is a lack of a foolproof system for making early sepsis diagnosis because conventional diagnostic tools like cultures take a long time and are often burdened with false negatives, while molecular techniques require specific equipment and have high costs. In this context, biomarkers, such as C-Reactive Protein (CRP) and Procalcitonin (PCT), are very useful tools to distinguish between normal and pathological conditions, graduate the disease severity, guide treatment, monitor therapeutic responses and predict prognosis. Among the new emerging biomarkers of sepsis, Presepsin (P-SEP) appears to be the most promising. Several studies have shown that P-SEP plasma levels increase during bacterial sepsis and decline in response to appropriate therapy, with sensitivity and specificity values comparable to those of PCT. In neonatal sepsis, P-SEP compared to PCT has been shown to be more effective in diagnosing and guiding therapy. Since in sepsis the P-SEP plasma levels increase before those of PCT and since the current methods available allow measurement of P-SEP plasma levels within 17 min, P-SEP appears a sepsis biomarker particularly suited to the emergency department and critical care.
Collapse
Affiliation(s)
- Andrea Piccioni
- Emergency Medicine Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.P.); (S.C.); (M.M.P.); (M.C.); (M.C.); (F.F.)
| | - Michele Cosimo Santoro
- Emergency Medicine Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.P.); (S.C.); (M.M.P.); (M.C.); (M.C.); (F.F.)
- Correspondence:
| | - Tommaso de Cunzo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.d.C.); (G.T.); (A.S.); (F.V.)
| | - Gianluca Tullo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.d.C.); (G.T.); (A.S.); (F.V.)
| | - Sara Cicchinelli
- Emergency Medicine Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.P.); (S.C.); (M.M.P.); (M.C.); (M.C.); (F.F.)
| | - Angela Saviano
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.d.C.); (G.T.); (A.S.); (F.V.)
| | - Federico Valletta
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.d.C.); (G.T.); (A.S.); (F.V.)
| | - Marco Maria Pascale
- Emergency Medicine Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.P.); (S.C.); (M.M.P.); (M.C.); (M.C.); (F.F.)
| | - Marcello Candelli
- Emergency Medicine Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.P.); (S.C.); (M.M.P.); (M.C.); (M.C.); (F.F.)
| | - Marcello Covino
- Emergency Medicine Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.P.); (S.C.); (M.M.P.); (M.C.); (M.C.); (F.F.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.d.C.); (G.T.); (A.S.); (F.V.)
| | - Francesco Franceschi
- Emergency Medicine Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.P.); (S.C.); (M.M.P.); (M.C.); (M.C.); (F.F.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (T.d.C.); (G.T.); (A.S.); (F.V.)
| |
Collapse
|
13
|
Non-Immunotherapy Application of LNP-mRNA: Maximizing Efficacy and Safety. Biomedicines 2021; 9:biomedicines9050530. [PMID: 34068715 PMCID: PMC8151051 DOI: 10.3390/biomedicines9050530] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 02/07/2023] Open
Abstract
Lipid nanoparticle (LNP) formulated messenger RNA-based (LNP-mRNA) vaccines came into the spotlight as the first vaccines against SARS-CoV-2 virus to be applied worldwide. Long-known benefits of mRNA-based technologies consisting of relatively simple and fast engineering of mRNA encoding for antigens and proteins of interest, no genomic integration, and fast and efficient manufacturing process compared with other biologics have been verified, thus establishing a basis for a broad range of applications. The intrinsic immunogenicity of LNP formulated in vitro transcribed (IVT) mRNA is beneficial to the LNP-mRNA vaccines. However, avoiding immune activation is critical for therapeutic applications of LNP-mRNA for protein replacement where targeted mRNA expression and repetitive administration of high doses for a lifetime are required. This review summarizes our current understanding of immune activation induced by mRNA, IVT byproducts, and LNP. It gives a comprehensive overview of the present status of preclinical and clinical studies in which LNP-mRNA is used for protein replacement and treatment of rare diseases with an emphasis on safety. Moreover, the review outlines innovations and strategies to advance pharmacology and safety of LNP-mRNA for non-immunotherapy applications.
Collapse
|
14
|
Cationic Nanoparticle-Based Cancer Vaccines. Pharmaceutics 2021; 13:pharmaceutics13050596. [PMID: 33919378 PMCID: PMC8143365 DOI: 10.3390/pharmaceutics13050596] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 12/15/2022] Open
Abstract
Cationic nanoparticles have been shown to be surprisingly effective as cancer vaccine vehicles in preclinical and clinical studies. Cationic nanoparticles deliver tumor-associated antigens to dendritic cells and induce immune activation, resulting in strong antigen-specific cellular immune responses, as shown for a wide variety of vaccine candidates. In this review, we discuss the relation between the cationic nature of nanoparticles and the efficacy of cancer immunotherapy. Multiple types of lipid- and polymer-based cationic nanoparticulate cancer vaccines with various antigen types (e.g., mRNA, DNA, peptides and proteins) and adjuvants are described. Furthermore, we focus on the types of cationic nanoparticles used for T-cell induction, especially in the context of therapeutic cancer vaccination. We discuss different cationic nanoparticulate vaccines, molecular mechanisms of adjuvanticity and biodistribution profiles upon administration via different routes. Finally, we discuss the perspectives of cationic nanoparticulate vaccines for improving immunotherapy of cancer.
Collapse
|
15
|
Atho'illah MF, Safitri YD, Nur'aini FD, Widyarti S, Tsuboi H, Rifa'i M. Elicited soybean extract attenuates proinflammatory cytokines expression by modulating TLR3/TLR4 activation in high-fat, high-fructose diet mice. J Ayurveda Integr Med 2021; 12:43-51. [PMID: 33531194 PMCID: PMC8039419 DOI: 10.1016/j.jaim.2021.01.003] [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] [Received: 11/11/2020] [Revised: 12/31/2020] [Accepted: 01/08/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The high-fat, high-fructose diet (HFFD) provokes overnutrition and inflammation directly, mainly through Toll-like receptors (TLRs). Soybean (Glycine max L.) contains isoflavone that can be transformed into glyceollin by microbial and physical stimuli. Glyceollin possesses many beneficial effects on health. OBJECTIVE This study evaluates the beneficial effect of soybean extract elicited by Saccharomyces cerevisiae and light (ESE) on dendritic cells (DCs) profile and naïve T cells in HFFD mice. MATERIALS AND METHODS Female Balb/C mice were fed with HFFD for 24 weeks then orally administered with simvastatin 2.8 mg/kg BW or ESE 78, 104, and 130 mg/kg BW at the last four weeks. The expression of splenic CD11c+TLR3+, CD11c+TLR4+, NFκB+, CD11c+IL-17+, CD11c+TNF-α+, CD4+CD62L+, and CD8+CD62L+ subsets was measured by flow cytometry. The molecular docking has been measured using Pyrx 0.8, displayed in PyMol and Biovia Discovery Studio. RESULT HFFD significantly increased CD11c+TLR3+, CD11c+TLR4+, NFκB+, CD11c+IL-17+, CD11c+TNF-α+ expression and decreased CD4+CD62L+ and CD8+CD62L+ (p < 0.05) compared to normal diet (ND) groups. ESE reduced CD11c+TLR3+, CD11c+TLR4+, thereby decreasing NFκB+, as well as decreased the CD11c+IL-17+, CD11c+TNF-α+, and restores CD4+CD62L+ and CD8+CD62L+ subsets in HFFD mice. Glyceollin II exhibited the best binding affinity with an average energy of -7.3 kcal/mol to TLR3 and -7.9 kcal/mol to TLR4. CONCLUSION The bioactive compound in ESE act synergistically to modulate TLR3/TLR4 activation, reduced NFκB, IL-17, and TNF-α, and restores naïve T cells expression in HFFD mice. ESE was a favorable candidate to mitigate chronic inflammation.
Collapse
Affiliation(s)
- Mochammad Fitri Atho'illah
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, 65145, Malang, East Java, Indonesia
| | - Yunita Diyah Safitri
- Medical Laboratory Technology Program, Sekolah Tinggi Ilmu Kesehatan Karya Putra Bangsa, 66291, Tulungagung, East Java, Indonesia
| | - Farida Dewi Nur'aini
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, 65145, Malang, East Java, Indonesia
| | - Sri Widyarti
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, 65145, Malang, East Java, Indonesia
| | - Hideo Tsuboi
- Department of Immunology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showaku, Nagoya, 466-8550, Japan
| | - Muhaimin Rifa'i
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, 65145, Malang, East Java, Indonesia; Center of Biosystem Study, LPPM of Brawijaya University, 65145, Malang, East Java, Indonesia.
| |
Collapse
|
16
|
Ruiz‐Hitzky E, Darder M, Wicklein B, Ruiz‐Garcia C, Martín‐Sampedro R, del Real G, Aranda P. Nanotechnology Responses to COVID-19. Adv Healthc Mater 2020; 9:e2000979. [PMID: 32885616 DOI: 10.1002/adhm.202000979] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/24/2020] [Indexed: 12/12/2022]
Abstract
Researchers, engineers, and medical doctors are made aware of the severity of the COVID-19 infection and act quickly against the coronavirus SARS-CoV-2 using a large variety of tools. In this review, a panoply of nanoscience and nanotechnology approaches show how these disciplines can help the medical, technical, and scientific communities to fight the pandemic, highlighting the development of nanomaterials for detection, sanitation, therapies, and vaccines. SARS-CoV-2, which can be regarded as a functional core-shell nanoparticle (NP), can interact with diverse materials in its vicinity and remains attached for variable times while preserving its bioactivity. These studies are critical for the appropriate use of controlled disinfection systems. Other nanotechnological approaches are also decisive for the development of improved novel testing and diagnosis kits of coronavirus that are urgently required. Therapeutics are based on nanotechnology strategies as well and focus on antiviral drug design and on new nanoarchitectured vaccines. A brief overview on patented work is presented that emphasizes nanotechnology applied to coronaviruses. Finally, some comments are made on patents of the initial technological responses to COVID-19 that have already been put in practice.
Collapse
Affiliation(s)
- Eduardo Ruiz‐Hitzky
- Materials Science Institute of Madrid ICMM‐CSIC c/ Sor Juana Inés de la Cruz 3 Madrid 28049 Spain
| | - Margarita Darder
- Materials Science Institute of Madrid ICMM‐CSIC c/ Sor Juana Inés de la Cruz 3 Madrid 28049 Spain
| | - Bernd Wicklein
- Materials Science Institute of Madrid ICMM‐CSIC c/ Sor Juana Inés de la Cruz 3 Madrid 28049 Spain
| | | | - Raquel Martín‐Sampedro
- Materials Science Institute of Madrid ICMM‐CSIC c/ Sor Juana Inés de la Cruz 3 Madrid 28049 Spain
- National Institute of Agricultural and Food Research INIA Ctra. de la Coruña Km 7.5 Madrid 28040 Spain
| | - Gustavo del Real
- National Institute of Agricultural and Food Research INIA Ctra. de la Coruña Km 7.5 Madrid 28040 Spain
| | - Pilar Aranda
- Materials Science Institute of Madrid ICMM‐CSIC c/ Sor Juana Inés de la Cruz 3 Madrid 28049 Spain
| |
Collapse
|
17
|
Lipid Nanoparticle Acts as a Potential Adjuvant for Influenza Split Vaccine without Inducing Inflammatory Responses. Vaccines (Basel) 2020; 8:vaccines8030433. [PMID: 32756368 PMCID: PMC7565178 DOI: 10.3390/vaccines8030433] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 12/13/2022] Open
Abstract
Vaccination is a critical and reliable strategy for controlling the spread of influenza viruses in populations. Conventional seasonal split vaccines (SVs) for influenza evoke weaker immune responses than other types of vaccines, such as inactivated whole-virion vaccines, although SVs are highly safe compared to other types. Here, we assessed the potential of the lipid nanoparticle (LNP) we developed as an adjuvant for conventional influenza SV as an antigen in mice. The LNP did not induce the production of cytokines such as interleukin-6 (IL-6) and IL-12 p40 by dendritic cells or the expression of co-stimulatory molecules on these cells in vitro. In contrast, an SV adjuvanted with LNP improved SV-specific IgG1 and IgG2 responses and the Th1 response compared to the SV alone in mice. In addition, SV adjuvanted with an LNP gave superior protection against the influenza virus challenge over the SV alone and was as effective as SV adjuvanted with aluminum salts in mice. The LNP did not provoke inflammatory responses such as inflammatory cytokine production and inflammatory immune cell infiltration in mice, whereas aluminum salts induced inflammatory responses. These results suggest the potential of the LNP as an adjuvant without inflammatory responses for influenza SVs. Our strategy should be useful for developing influenza vaccines with enhanced efficacy and safety.
Collapse
|
18
|
Shetab Boushehri MA, Dietrich D, Lamprecht A. Nanotechnology as a Platform for the Development of Injectable Parenteral Formulations: A Comprehensive Review of the Know-Hows and State of the Art. Pharmaceutics 2020; 12:pharmaceutics12060510. [PMID: 32503171 PMCID: PMC7356945 DOI: 10.3390/pharmaceutics12060510] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
Within recent decades, the development of nanotechnology has made a significant contribution to the progress of various fields of study, including the domains of medical and pharmaceutical sciences. A substantially transformed arena within the context of the latter is the development and production of various injectable parenteral formulations. Indeed, recent decades have witnessed a rapid growth of the marketed and pipeline nanotechnology-based injectable products, which is a testimony to the remarkability of the aforementioned contribution. Adjunct to the ability of nanomaterials to deliver the incorporated payloads to many different targets of interest, nanotechnology has substantially assisted to the development of many further facets of the art. Such contributions include the enhancement of the drug solubility, development of long-acting locally and systemically injectable formulations, tuning the onset of the drug’s release through the endowment of sensitivity to various internal or external stimuli, as well as adjuvancy and immune activation, which is a desirable component for injectable vaccines and immunotherapeutic formulations. The current work seeks to provide a comprehensive review of all the abovementioned contributions, along with the most recent advances made within each domain. Furthermore, recent developments within the domains of passive and active targeting will be briefly debated.
Collapse
Affiliation(s)
- Maryam A. Shetab Boushehri
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- Correspondence: ; Tel.: +49-228-736428; Fax: +49-228-735268
| | - Dirk Dietrich
- Department of Neurosurgery, University Clinic of Bonn, 53105 Bonn, Germany;
| | - Alf Lamprecht
- Department of Pharmaceutics, Faculty of Pharmacy, University of Bonn, 53121 Bonn, Germany;
- PEPITE EA4267, Institute of Pharmacy, University Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
19
|
Khan MAA, Ami JQ, Faisal K, Chowdhury R, Ghosh P, Hossain F, Abd El Wahed A, Mondal D. An immunoinformatic approach driven by experimental proteomics: in silico design of a subunit candidate vaccine targeting secretory proteins of Leishmania donovani amastigotes. Parasit Vectors 2020; 13:196. [PMID: 32295617 PMCID: PMC7160903 DOI: 10.1186/s13071-020-04064-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 04/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background Visceral leishmaniasis (VL) caused by dimorphic Leishmania species is a parasitic disease with high socioeconomic burden in endemic areas worldwide. Sustaining control of VL in terms of proper and prevailing immunity development is a global necessity amid unavailability of a prophylactic vaccine. Screening of experimental proteome of the human disease propagating form of Leishmania donovani (amastigote) can be more pragmatic for in silico mining of novel vaccine candidates. Methods By using an immunoinformatic approach, CD4+ and CD8+ T cell-specific epitopes from experimentally reported L. donovani proteins having secretory potential and increased abundance in amastigotes were screened. A chimera linked with a Toll-like receptor 4 (TLR4) peptide adjuvant was constructed and evaluated for physicochemical characteristics, binding interaction with TLR4 in simulated physiological condition and the trend of immune response following hypothetical immunization. Results Selected epitopes from physiologically important L. donovani proteins were found mostly conserved in L. infantum, covering theoretically more than 98% of the global population. The multi-epitope chimeric vaccine was predicted as stable, antigenic and non-allergenic. Structural analysis of vaccine-TLR4 receptor docked complex and its molecular dynamics simulation suggest sufficiently stable binding interface along with prospect of non-canonical receptor activation. Simulation dynamics of immune response following hypothetical immunization indicate active and memory B as well as CD4+ T cell generation potential, and likely chance of a more Th1 polarized response. Conclusions The methodological approach and results from this study could facilitate more informed screening and selection of candidate antigenic proteins for entry into vaccine production pipeline in future to control human VL.![]()
Collapse
Affiliation(s)
- Md Anik Ashfaq Khan
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, 1212, Bangladesh
| | - Jenifar Quaiyum Ami
- Infectious Diseases Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, 1212, Bangladesh
| | - Khaledul Faisal
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, 1212, Bangladesh
| | - Rajashree Chowdhury
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, 1212, Bangladesh
| | - Prakash Ghosh
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, 1212, Bangladesh
| | - Faria Hossain
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, 1212, Bangladesh
| | - Ahmed Abd El Wahed
- Microbiology and Animal Hygiene Division, Georg-August-University Goettingen, Burckhardtweg 2, 37077, Göttingen, Germany.
| | - Dinesh Mondal
- Nutrition and Clinical Services Division, International Centre for Diarrheal Disease Research, Bangladesh, Dhaka, 1212, Bangladesh.
| |
Collapse
|
20
|
Afroz R, Zhou Y, Little PJ, Xu S, Mohamed R, Stow J, Kamato D. Toll-like Receptor 4 Stimulates Gene Expression via Smad2 Linker Region Phosphorylation in Vascular Smooth Muscle Cells. ACS Pharmacol Transl Sci 2020; 3:524-534. [PMID: 32566917 DOI: 10.1021/acsptsci.9b00113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Indexed: 02/06/2023]
Abstract
Atherosclerosis begins in the vessel wall with the retention of low density lipoproteins to modified proteoglycans with hyperelongated glycosaminoglycan (GAG) chains. Bacterial infections produce endotoxins such as lipopolysaccharide that exacerbate the outcome of atherosclerosis by generating a heightened state of inflammation. Lipopolysaccharide (LPS) via its toll-like receptor (TLR) is well-known for its role in mediating an inflammatory response in the body. Emerging evidence demonstrates that TLRs are involved in regulating vascular functions. In this study we sought to investigate the role of LPS in proteoglycan modification and GAG chain elongation, and we hypothesize that LPS will signal via Smad2 dependent pathways to regulate GAG chain elongation. The in vitro model used human aortic vascular smooth muscle cells. GAG gene expression was assessed by quantitative real-time polymerase chain reaction. Western blotting was performed using whole-cell protein lysates to assess the signaling pathway. LPS via TLR4 stimulates the expression of GAG synthesizing enzymes to an equal extent to traditional cardiovascular agonists. LPS phosphorylates the Smad2 linker region via TAK-1/MAPK dependent pathways which correlated with genes associated with GAG chain initiation and elongation. The well-characterized role of LPS in inflammation and our data on GAG gene expression demonstrates that GAG chain elongation is the earliest marker of the inflammatory cascade in atherosclerosis development.
Collapse
Affiliation(s)
- Rizwana Afroz
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Ying Zhou
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| | - Suowen Xu
- Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Raafat Mohamed
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Jennifer Stow
- Institute of Molecular Bioscience, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| |
Collapse
|
21
|
Pizzuto M, Lonez C, Baroja-Mazo A, Martínez-Banaclocha H, Tourlomousis P, Gangloff M, Pelegrin P, Ruysschaert JM, Gay NJ, Bryant CE. Saturation of acyl chains converts cardiolipin from an antagonist to an activator of Toll-like receptor-4. Cell Mol Life Sci 2019; 76:3667-3678. [PMID: 31062071 PMCID: PMC6697720 DOI: 10.1007/s00018-019-03113-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/12/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
Cardiolipins (CLs) are tetra-acylated diphosphatidylglycerols found in bacteria, yeast, plants, and animals. In healthy mammals, CLs are unsaturated, whereas saturated CLs are found in blood cells from Barth syndrome patients and in some Gram-positive bacteria. Here, we show that unsaturated but not saturated CLs block LPS-induced NF-κB activation, TNF-α and IP-10 secretion in human and murine macrophages, as well as LPS-induced TNF-α and IL-1β release in human blood mononuclear cells. Using HEK293 cells transfected with Toll-like receptor 4 (TLR4) and its co-receptor Myeloid Differentiation 2 (MD2), we demonstrate that unsaturated CLs compete with LPS for binding TLR4/MD2 preventing its activation, whereas saturated CLs are TLR4/MD2 agonists. As a consequence, saturated CLs induce a pro-inflammatory response in macrophages characterized by TNF-α and IP-10 secretion, and activate the alternative NLRP3 inflammasome pathway in human blood-derived monocytes. Thus, we identify that double bonds discriminate between anti- and pro-inflammatory properties of tetra-acylated molecules, providing a rationale for the development of TLR4 activators and inhibitors for use as vaccine adjuvants or in the treatment of TLR4-related diseases.
Collapse
Affiliation(s)
- Malvina Pizzuto
- Structure and Function of Biological Membranes, Université Libre de Bruxelles, Blvd du Triomphe Access 2, 1050, Brussels, Belgium.
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK.
- Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge, CB3 0ES, UK.
- Molecular Inflammation Group, Biomedical Research Institute of Murcia IMIB-Arrixaca, Clinical University Hospital Virgen de la Arrixaca, Carretera Buenavista s/n, 30120, Murcia, Spain.
| | - Caroline Lonez
- Structure and Function of Biological Membranes, Université Libre de Bruxelles, Blvd du Triomphe Access 2, 1050, Brussels, Belgium
- Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge, CB3 0ES, UK
| | - Alberto Baroja-Mazo
- Molecular Inflammation Group, Biomedical Research Institute of Murcia IMIB-Arrixaca, Clinical University Hospital Virgen de la Arrixaca, Carretera Buenavista s/n, 30120, Murcia, Spain
| | - Helios Martínez-Banaclocha
- Molecular Inflammation Group, Biomedical Research Institute of Murcia IMIB-Arrixaca, Clinical University Hospital Virgen de la Arrixaca, Carretera Buenavista s/n, 30120, Murcia, Spain
| | - Panagiotis Tourlomousis
- Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge, CB3 0ES, UK
| | - Monique Gangloff
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Pablo Pelegrin
- Molecular Inflammation Group, Biomedical Research Institute of Murcia IMIB-Arrixaca, Clinical University Hospital Virgen de la Arrixaca, Carretera Buenavista s/n, 30120, Murcia, Spain
| | - Jean-Marie Ruysschaert
- Structure and Function of Biological Membranes, Université Libre de Bruxelles, Blvd du Triomphe Access 2, 1050, Brussels, Belgium
| | - Nicholas J Gay
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, CB2 1GA, UK
| | - Clare E Bryant
- Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge, CB3 0ES, UK
| |
Collapse
|
22
|
Antibody responses to crucial functional epitopes as a novel approach to assess immunogenicity of vaccine adjuvants. Vaccine 2019; 37:3770-3778. [PMID: 31153687 DOI: 10.1016/j.vaccine.2019.05.068] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/08/2019] [Accepted: 05/22/2019] [Indexed: 12/22/2022]
Abstract
We are interested in developing a vaccine that prevents genital herpes. Adjuvants have a major impact on vaccine immunogenicity. We compared two adjuvants, an experimental Merck Sharp & Dohme lipid nanoparticle (LNP) adjuvant, LNP-2, with CpG oligonucleotide combined with alum for immunogenicity in mice when administered with herpes simplex virus type 2 (HSV-2) glycoproteins C, D and E (gC2, gD2, gE2). The immunogens are intended to produce neutralizing antibodies to gC2 and gD2, antibodies to gD2 and gE2 that block cell-to-cell spread, and antibodies to gE2 and gC2 that block immune evasion from antibody and complement, respectively. Overall, CpG/alum was better at producing serum and vaginal IgG binding antibodies, neutralizing antibodies, antibodies that block virus spread from cell-to-cell, and antibodies that block immune evasion domains on gC2. We used a novel high throughput biosensor assay to further assess differences in immunogenicity by mapping antibody responses to seven crucial epitopes on gD2 involved in virus entry or cell-to-cell spread. We found striking differences between CpG/alum and LNP-2. Mice immunized with gD2 CpG/alum produced higher titers of antibodies than LNP-2 to six of seven crucial epitopes and produced antibodies to more crucial epitopes than LNP-2. Measuring epitope-specific antibodies helped to define mechanisms by which CpG/alum outperformed LNP-2 and is a valuable technique to compare adjuvants.
Collapse
|
23
|
Gandhapudi SK, Ward M, Bush JPC, Bedu-Addo F, Conn G, Woodward JG. Antigen Priming with Enantiospecific Cationic Lipid Nanoparticles Induces Potent Antitumor CTL Responses through Novel Induction of a Type I IFN Response. THE JOURNAL OF IMMUNOLOGY 2019; 202:3524-3536. [PMID: 31053626 DOI: 10.4049/jimmunol.1801634] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/14/2019] [Indexed: 02/06/2023]
Abstract
Certain types of cationic lipids have shown promise in cancer immunotherapy, but their mechanism of action is poorly understood. In this study, we describe the properties of an immunotherapeutic consisting of the pure cationic lipid enantiomer R-1,2-dioleoyl-3-trimethyl-ammonium-propane (R-DOTAP) formulated with modified viral or self-peptide Ags. R-DOTAP formulations with peptide Ags stimulate strong cross-presentation and potent CD8 T cell responses associated with a high frequency of polyfunctional CD8 T cells. In a human papillomavirus tumor model system, a single s.c. injection of tumor-bearing mice with R-DOTAP plus human papillomavirus Ags induces complete regression of large tumors associated with an influx of Ag-specific CD8 T cells and a reduction of the ratio of regulatory/Ag-specific CD8 T cells. R-DOTAP also synergizes with an anti-PD1 checkpoint inhibitor, resulting in a significant inhibition of B16 melanoma tumor growth. We found that R-DOTAP stimulates type I IFN production by dendritic cells in vivo and in vitro. s.c. injection of R-DOTAP results in an IFN-dependent increase in draining lymph node size and a concomitant increase in CD69 expression. Using knockout mice, we show that type I IFN is required for the induction of CD8 T cell activity following administration of R-DOTAP plus Ag. This response requires Myd88 but not TRIF or STING. We also show that R-DOTAP stimulates both TLR7 and 9. Collectively, these studies reveal that R-DOTAP stimulates endosomal TLRs, resulting in a Myd88-dependent production of type I IFN. When administered with Ag, this results in potent Ag-specific CD8 T cell responses and antitumor activity.
Collapse
Affiliation(s)
- Siva K Gandhapudi
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536; and
| | - Martin Ward
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536; and
| | - John Peyton C Bush
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536; and
| | | | - Greg Conn
- PDS Biotechnology Corporation, Princeton, NJ 08540
| | - Jerold G Woodward
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536; and
| |
Collapse
|
24
|
Grandellis C, Garavaglia BS, Gottig N, Lonez C, Ruysschaert JM, Ottado J. DOTAP, a lipidic transfection reagent, triggers Arabidopsis plant defense responses. PLANTA 2019; 249:469-480. [PMID: 30255355 DOI: 10.1007/s00425-018-3014-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 09/20/2018] [Indexed: 06/08/2023]
Abstract
DOTAP triggers Arabidopsis thaliana immunity and by priming the defense response is able to reduce bacterial pathogen attack. DOTAP is a cationic lipid widely used as a liposomal transfection reagent and it has recently been identified as a strong activator of the innate immune system in animal cells. Plants are sessile organisms and unlike mammals, that have innate and acquired immunity, plants possess only innate immunity. A key feature of plant immunity is the ability to sense potentially dangerous signals, as it is the case for microbe-associated, pathogen-associated or damage-associated molecular patterns and by doing so, trigger an active defense response to cope with the perturbing stimulus. Here, we evaluated the effect of DOTAP in plant basal innate immunity. An initial plant defense response was induced by the cationic lipid DOTAP in the model plant Arabidopsis thaliana, assessed by callose deposition, reactive oxygen species production, and plant cell death. In addition, a proteomic analysis revealed that these responses are mirrored by changes in the plant proteome, such as up-regulation of proteins related to defense responses, including proteins involved in photorespiration, cysteine and oxylipin synthesis, and oxidative stress response; and down-regulation of enzymes related to photosynthesis. Furthermore, DOTAP was able to prime the defense response for later pathogenic challenges as in the case of the virulent bacterial pathogen Pseudomonas syringae pv. tomato. Disease outcome was diminished in DOTAP-pre-treated leaves and bacterial growth was reduced 100 times compared to mock leaves. Therefore, DOTAP may be considered a good candidate as an elicitor for the study of plant immunity.
Collapse
Affiliation(s)
- Carolina Grandellis
- Instituto de Biología Molecular Y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas (IBR-CONICET) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Ocampo y Esmeralda, 2000, Rosario, Argentina
| | - Betiana S Garavaglia
- Instituto de Biología Molecular Y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas (IBR-CONICET) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Ocampo y Esmeralda, 2000, Rosario, Argentina
| | - Natalia Gottig
- Instituto de Biología Molecular Y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas (IBR-CONICET) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Ocampo y Esmeralda, 2000, Rosario, Argentina
| | - Caroline Lonez
- Laboratory of Structure and Function of Biological Membranes, Université Libre de Bruxelles, Boulevard du Triomphe, 1050, Brussels, Belgium
| | - Jean-Marie Ruysschaert
- Laboratory of Structure and Function of Biological Membranes, Université Libre de Bruxelles, Boulevard du Triomphe, 1050, Brussels, Belgium
| | - Jorgelina Ottado
- Instituto de Biología Molecular Y Celular de Rosario, Consejo Nacional de Investigaciones Científicas y Técnicas (IBR-CONICET) and Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Ocampo y Esmeralda, 2000, Rosario, Argentina.
| |
Collapse
|
25
|
Memar MY, Baghi HB. Presepsin: A promising biomarker for the detection of bacterial infections. Biomed Pharmacother 2019; 111:649-656. [PMID: 30611989 DOI: 10.1016/j.biopha.2018.12.124] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 12/28/2018] [Accepted: 12/30/2018] [Indexed: 12/22/2022] Open
Abstract
Appropriate recognition of bacterial infections in health care setting is the basis for effective treatment and control of infectious diseases. The positivity rate of traditional methods is low and is influenced by quality and quantity of specimens, patient antibiotic administration, severity of infection, and laboratory sufficiency. Currently, there are novel non-culture-based techniques that are being accomplished to improve the identification of infections. Several immunologic biomarkers have been assessed to develop the best indicator of infections. Presepsin is an immunologic biomarker which has been demonstrated as new, emerging, early indicator for the detection of different infections. The biological function of presepsin is not well known. However, it is believed that it may be a regulatory molecule of the adaptive immune system and also a stimulator of monocyte phagocytosis. The early increased levels of presepsin during the sepsis and other bacterial infections have made it an attractive indicator for laboratory testing. Several studies have investigated the capacity of presepsin for use in clinical settings. The aim of the present study was review the clinical application of presepsin in diagnosis and prediction of infections. To achieve this objective, the documents on diagnostic and clinical assessment were evaluated in PubMed and Scopus databases regarding the use of presepsin as indicators of infections.
Collapse
Affiliation(s)
- Mohammad Yousef Memar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
26
|
Shetab Boushehri MA, Lamprecht A. TLR4-Based Immunotherapeutics in Cancer: A Review of the Achievements and Shortcomings. Mol Pharm 2018; 15:4777-4800. [DOI: 10.1021/acs.molpharmaceut.8b00691] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, D-53121 Bonn, Germany
- PEPITE EA4267, Univ. Bourgonge Franch-Comte, 25030 Besançon, France
| |
Collapse
|
27
|
Pizzuto M, Bigey P, Lachagès AM, Hoffmann C, Ruysschaert JM, Escriou V, Lonez C. Cationic lipids as one-component vaccine adjuvants: A promising alternative to alum. J Control Release 2018; 287:67-77. [PMID: 30110615 DOI: 10.1016/j.jconrel.2018.08.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 07/23/2018] [Accepted: 08/10/2018] [Indexed: 12/19/2022]
Abstract
Effective vaccine formulations consist of several components: an antigen carrier, the antigen, a stimulator of cellular immunity such as a Toll-like Receptors (TLRs) ligand, and a stimulator of humoral response such as an inflammasome activator. Here, we investigated the immunostimulatory and adjuvant properties of lipopolyamines, cationic lipids used as gene carriers. We identified new lipopolyamines able to activate both TLR2 and TLR4 and showed that lipopolyamines interact with TLRs via a mechanism different from the one used by bacterial ligands, activating a strong type-I IFN response, pro-inflammatory cytokines and IL-1β secretion. The TLR and inflammasome stimulations, together with the antigen carrier properties of lipopolyamines, resulted in both humoral and cellular immunity in mice vaccinated against OVA and make lipopolyamines promising one-component vaccine adjuvants.
Collapse
Affiliation(s)
- Malvina Pizzuto
- Structure and Fonction of Biological Membranes, Université Libre de Bruxelles, Boulevard du Triomphe, 1050 Brussels, Belgium.
| | - Pascal Bigey
- CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), UMR 8258, F-75006 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité University, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, UTCBS, F-75005 Paris, France
| | - Anne-Marie Lachagès
- CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), UMR 8258, F-75006 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité University, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, UTCBS, F-75005 Paris, France
| | - Céline Hoffmann
- CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), UMR 8258, F-75006 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité University, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, UTCBS, F-75005 Paris, France
| | - Jean-Marie Ruysschaert
- Structure and Fonction of Biological Membranes, Université Libre de Bruxelles, Boulevard du Triomphe, 1050 Brussels, Belgium
| | - Virginie Escriou
- CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), UMR 8258, F-75006 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité University, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, UTCBS, F-75005 Paris, France
| | - Caroline Lonez
- Structure and Fonction of Biological Membranes, Université Libre de Bruxelles, Boulevard du Triomphe, 1050 Brussels, Belgium; Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge CB3 0ES, United Kingdom
| |
Collapse
|
28
|
Pizzuto M, Gangloff M, Scherman D, Gay NJ, Escriou V, Ruysschaert JM, Lonez C. Toll-like receptor 2 promiscuity is responsible for the immunostimulatory activity of nucleic acid nanocarriers. J Control Release 2016; 247:182-193. [PMID: 28040465 PMCID: PMC5312493 DOI: 10.1016/j.jconrel.2016.12.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 12/23/2016] [Indexed: 12/14/2022]
Abstract
Lipopolyamines (LPAs) are cationic lipids; they interact spontaneously with nucleic acids to form lipoplexes used for gene delivery. The main hurdle to using lipoplexes in gene therapy lies in their immunostimulatory properties, so far attributed to the nucleic acid cargo, while cationic lipids were considered as inert to the immune system. Here we demonstrate for the first time that di-C18 LPAs trigger pro-inflammatory responses through Toll-like receptor 2 (TLR2) activation, and this whether they are bound to nucleic acids or not. Molecular docking experiments suggest potential TLR2 binding modes reminiscent of bacterial lipopeptide sensing. The di-C18 LPAs share the ability of burying their lipid chains in the hydrophobic cavity of TLR2 and, in some cases, TLR1, at the vicinity of the dimerization interface; the cationic headgroups form multiple hydrogen bonds, thus crosslinking TLRs into functional complexes. Unravelling the molecular basis of TLR1 and TLR6-driven heterodimerization upon LPA binding underlines the highly collaborative and promiscuous ligand binding mechanism. The prevalence of non-specific main chain-mediated interactions demonstrates that potentially any saturated LPA currently used or proposed as transfection agent is likely to activate TLR2 during transfection. Hence our study emphasizes the urgent need to test the inflammatory properties of transfection agents and proposes the use of docking analysis as a preliminary screening tool for the synthesis of new non-immunostimulatory nanocarriers.
Collapse
Affiliation(s)
- Malvina Pizzuto
- Structure and Function of Biological Membranes, Université Libre de Bruxelles, Boulevard du Triomphe, 1050 Brussels, Belgium.
| | - Monique Gangloff
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, UK.
| | - Daniel Scherman
- CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), UMR 8258, F-75006 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité University, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, UTCBS, F-75005 Paris, France
| | - Nicholas J Gay
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge, UK
| | - Virginie Escriou
- CNRS, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), UMR 8258, F-75006 Paris, France; INSERM, UTCBS U 1022, F-75006 Paris, France; Université Paris Descartes, Sorbonne-Paris-Cité University, UTCBS, F-75006 Paris, France; Chimie ParisTech, PSL Research University, UTCBS, F-75005 Paris, France
| | - Jean-Marie Ruysschaert
- Structure and Function of Biological Membranes, Université Libre de Bruxelles, Boulevard du Triomphe, 1050 Brussels, Belgium
| | - Caroline Lonez
- Structure and Function of Biological Membranes, Université Libre de Bruxelles, Boulevard du Triomphe, 1050 Brussels, Belgium; Department of Veterinary Medicine, University of Cambridge, Madingley Rd, Cambridge CB3 0ES, UK
| |
Collapse
|
29
|
Extracellular Vesicle Proteins Associated with Systemic Vascular Events Correlate with Heart Failure: An Observational Study in a Dyspnoea Cohort. PLoS One 2016; 11:e0148073. [PMID: 26820481 PMCID: PMC4731211 DOI: 10.1371/journal.pone.0148073] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/12/2016] [Indexed: 01/09/2023] Open
Abstract
Background SerpinF2, SerpinG1, CystatinC and CD14 are involved in inflammatory processes and plasma extracellular vesicle (EV) -levels of these proteins have been reported to be associated with systemic vascular events. Evidence is accumulating that inflammatory processes may play a pivotal role both in systemic vascular events and in heart failure. Therefore, we studied the association between plasma extracellular vesicle SerpinF2-, SerpinG1-, CystatinC and CD14-levels and the occurrence of acute heart failure in patients. Methods and Result Extracellular vesicle protein levels of SerpinG1, SerpinF2, CystatinC and CD14 were measured in an observational study of 404 subjects presenting with dysponea at the emergency department (4B-cohort). Plasma extracellular vesicles were precipitated in a total extracellular vesicles (TEX)-fraction and in separate LDL- and HDL-subfractions. Extracellular vesicle protein levels were measured with a quantitative immune assay in all 3 precipitates. Out of 404 subjects, 141 (35%) were diagnosed with acutely decompensated heart failure. After correction for confounders (including comorbidities and medications), levels of CD14 in the HDL-fraction (OR 1.53, p = 0.01), SerpinF2 in the TEX-and LDL-fraction (ORs respectively 0.71 and 0.65, p<0.05) and SerpinG1 in the TEX-fraction (OR 1.55, p = 0.004) were statistically significantly related to heart failure. Furthermore, extracellular vesicle CD14- and SerpinF2-levels were significantly higher in heart failure patients with preserved ejection fraction than in those with reduced ejection fraction. Conclusion Extracellular vesicle levels of CD14, SerpinG1 and SerpinF2 are associated with the occurrence of heart failure in subjects suspected for acute heart failure, suggesting common underlying pathophysiological mechanisms for heart failure and vascular events.
Collapse
|