1
|
Yoo S, Faisal M, Bae S, Youn K, Park H, Kwon SP, Hwang IK, Lee J, Kim HJ, Nam J, Keum G, Bang E. Novel Less Toxic, Lymphoid Tissue-Targeted Lipid Nanoparticles Containing a Vitamin B5-Derived Ionizable Lipid for mRNA Vaccine Delivery. Adv Healthc Mater 2025; 14:e2403366. [PMID: 39502027 PMCID: PMC11912100 DOI: 10.1002/adhm.202403366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/23/2024] [Indexed: 03/18/2025]
Abstract
Following their approval by the Food and Drug Administration, lipid nanoparticles (LNPs) have emerged as promising tools for delivering mRNA vaccines and therapeutics. Ionizable lipids are among the essential components of LNPs, as they play crucial roles in encapsulating mRNA and facilitating its release into the cytosol. In this study, 17 innovative ionizable lipids using vitamin B5 are designed as the core structure, aiming to reduce toxicity, to maintain vaccine efficiency, and to ensure synthetic feasibility. The top-performing LNP in terms of mRNA vaccine delivery in the mouse model is LNP 5097, which is generated by incorporating ionizable lipid I97. mRNA⊂LNP 5097 demonstrates favorable structural and physicochemical properties, high mRNA transfection efficiency, and long-term stability. Moreover, mRNA⊂LNP 5097 specifically delivers the mRNA to the spleen and lymph nodes in model mice, induces balanced Th1/Th2 immune responses, and elicits the production of high levels of neutralizing antibodies with low toxicity. The findings here suggest the high utility of LNP 5097, which includes novel vitamin B5-derived ionizable lipids with reduced toxicity, in mRNA vaccine research for both infectious diseases and cancer.
Collapse
Affiliation(s)
- Soyeon Yoo
- Brain Technology Research CenterBrain Science Research DivisionKorea Institute of Science and Technology5 Hwarang‐ro 14‐gil, Seongbuk‐guSeoul02792Republic of Korea
| | - Muhammad Faisal
- Brain Technology Research CenterBrain Science Research DivisionKorea Institute of Science and Technology5 Hwarang‐ro 14‐gil, Seongbuk‐guSeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
| | - Seo‐Hyeon Bae
- Department of Medical and Biological SciencesThe Catholic University of Korea43 Jibong‐roBucheonGyeonggi‐do14662Republic of Korea
- BK21 four Department of BiotechnologyThe Catholic University of Korea43 Jibong‐roBucheonGyeonggi‐do14662South Korea
| | - Kounghwa Youn
- Brain Technology Research CenterBrain Science Research DivisionKorea Institute of Science and Technology5 Hwarang‐ro 14‐gil, Seongbuk‐guSeoul02792Republic of Korea
- KHU‐KIST Department of Converging Science and TechnologyKyung Hee University7–13 Kyungheedae‐ro 6‐gil, Dongdaemun‐guSeoul02447Republic of Korea
| | - Hyo‐Jung Park
- Department of Medical and Biological SciencesThe Catholic University of Korea43 Jibong‐roBucheonGyeonggi‐do14662Republic of Korea
- BK21 four Department of BiotechnologyThe Catholic University of Korea43 Jibong‐roBucheonGyeonggi‐do14662South Korea
| | - Sung Pil Kwon
- Brain Technology Research CenterBrain Science Research DivisionKorea Institute of Science and Technology5 Hwarang‐ro 14‐gil, Seongbuk‐guSeoul02792Republic of Korea
| | - Il Kwon Hwang
- Brain Technology Research CenterBrain Science Research DivisionKorea Institute of Science and Technology5 Hwarang‐ro 14‐gil, Seongbuk‐guSeoul02792Republic of Korea
- Department of Chemical and Biological EngineeringKorea University145 Anam‐ro, Seongbuk‐guSeoul02841Republic of Korea
| | - Jisun Lee
- Department of Medical and Biological SciencesThe Catholic University of Korea43 Jibong‐roBucheonGyeonggi‐do14662Republic of Korea
| | - Hyeong Jun Kim
- Medicinal Materials Research CenterBiomedical Research DivisionKorea Institute of Science and Technology5 Hwarang‐ro 14‐gil, Seongbuk‐guSeoul02792Republic of Korea
| | - Jae‐Hwan Nam
- Department of Medical and Biological SciencesThe Catholic University of Korea43 Jibong‐roBucheonGyeonggi‐do14662Republic of Korea
- BK21 four Department of BiotechnologyThe Catholic University of Korea43 Jibong‐roBucheonGyeonggi‐do14662South Korea
- SML Biopharm, Gwangmyeong Station M Cluster17 Deokan‐ro 104beon‐gilGwangmyeong‐siGyeonggi‐do14353Republic of Korea
| | - Gyochang Keum
- Brain Technology Research CenterBrain Science Research DivisionKorea Institute of Science and Technology5 Hwarang‐ro 14‐gil, Seongbuk‐guSeoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolKorea University of Science and Technology (UST)Seoul02792Republic of Korea
| | - Eun‐Kyoung Bang
- Brain Technology Research CenterBrain Science Research DivisionKorea Institute of Science and Technology5 Hwarang‐ro 14‐gil, Seongbuk‐guSeoul02792Republic of Korea
- KHU‐KIST Department of Converging Science and TechnologyKyung Hee University7–13 Kyungheedae‐ro 6‐gil, Dongdaemun‐guSeoul02447Republic of Korea
- Department of Chemical and Biological EngineeringKorea University145 Anam‐ro, Seongbuk‐guSeoul02841Republic of Korea
| |
Collapse
|
2
|
Weth AF, Dangerfield EM, Timmer MSM, Stocker BL. Recent Advances in the Development of Mincle-Targeting Vaccine Adjuvants. Vaccines (Basel) 2024; 12:1320. [PMID: 39771982 PMCID: PMC11680293 DOI: 10.3390/vaccines12121320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 01/11/2025] Open
Abstract
The Macrophage-inducible C-type lectin (Mincle) is a pattern-recognition receptor (PRR), which has shown much promise as a molecular target for the development of TH1/TH17-skewing vaccine adjuvants. In 2009, the first non-proteinaceous Mincle ligands, trehalose dimycolate (TDM) and trehalose dibehenate (TDB), were identified. This prompted a search for other Mincle agonists and the exploration of Mincle agonists as vaccine adjuvants for both preventative and therapeutic (anti-cancer) vaccines. In this review, we discuss those classes of Mincle agonists that have been explored for their adjuvant potential. These Mincle agonists have been used as stand-alone adjuvants or in combination with other pathogen-associated molecular patterns (PAMPs) or immunomodulatory agents. We will also highlight recently identified Mincle ligands with hitherto unknown adjuvanticity. Conjugate vaccines that contain covalently linked adjuvants and/or adjuvant-antigen combinations are also presented, as well as the different formulations (e.g., oil-in-water emulsions, liposomes, and particulate delivery systems) that have been used for the codelivery of antigens and adjuvants. Insofar the reader is presented with a thorough review of the potential of Mincle-mediated vaccine adjuvants, including historical context, present-day research and clinical trials, and outstanding research questions, such as the role of ligand presentation and Mincle clustering, which, if better understood, will aid in the development of the much-needed TH1/TH17-skewing vaccine adjuvants.
Collapse
Affiliation(s)
| | | | - Mattie S. M. Timmer
- School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| | - Bridget L. Stocker
- School of Chemical and Physical Sciences, Victoria University of Wellington, P.O. Box 600, Wellington 6140, New Zealand
| |
Collapse
|
3
|
Noriega M, Corey RA, Haanappel E, Demange P, Czaplicki G, Atkinson RA, Chavent M. Coarse-Graining the Recognition of a Glycolipid by the C-Type Lectin Mincle Receptor. J Phys Chem B 2024; 128:9935-9946. [PMID: 39368102 DOI: 10.1021/acs.jpcb.4c03242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
Macrophage inducible Ca2+-dependent lectin (Mincle) receptor recognizes Mycobacterium tuberculosis glycolipids to trigger an immune response. This host membrane receptor is thus a key player in the modulation of the immune response to infection by M. tuberculosis and has emerged as a promising target for the development of new vaccines against tuberculosis. The recent development of the Martini 3 force field for coarse-grained (CG) molecular modeling allows the study of interactions of soluble proteins with small ligands which was not typically modeled well with the previous Martini 2 model. Here, we present a refined approach detailing a protocol for modeling interactions between a glycolipid and its receptor at a CG level using the Martini 3 force field. Using this approach, we studied Mincle and identified critical parameters governing ligand recognition, such as loop flexibility and the regulation of hydrophobic groove formation by calcium ions. In addition, we assessed ligand affinity using free energy perturbation calculations. Our results offer mechanistic insight into the interactions between Mincle and glycolipids, providing a basis for the rational design of molecules targeting this type of membrane receptors.
Collapse
Affiliation(s)
- Maxime Noriega
- Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR5089, CNRS-Université de Toulouse III-Paul Sabatier, BP 64182, 31077 Toulouse, Cedex 4, France
| | - Robin A Corey
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol BS8 1TD, U.K
| | - Evert Haanappel
- Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR5089, CNRS-Université de Toulouse III-Paul Sabatier, BP 64182, 31077 Toulouse, Cedex 4, France
- Laboratoire de Microbiologie et Génétique Moléculaires (LMGM), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31400, France
| | - Pascal Demange
- Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR5089, CNRS-Université de Toulouse III-Paul Sabatier, BP 64182, 31077 Toulouse, Cedex 4, France
| | - Georges Czaplicki
- Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR5089, CNRS-Université de Toulouse III-Paul Sabatier, BP 64182, 31077 Toulouse, Cedex 4, France
| | - R Andrew Atkinson
- Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR5089, CNRS-Université de Toulouse III-Paul Sabatier, BP 64182, 31077 Toulouse, Cedex 4, France
| | - Matthieu Chavent
- Institut de Pharmacologie et de Biologie Structurale (IPBS), UMR5089, CNRS-Université de Toulouse III-Paul Sabatier, BP 64182, 31077 Toulouse, Cedex 4, France
- Laboratoire de Microbiologie et Génétique Moléculaires (LMGM), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, Toulouse 31400, France
| |
Collapse
|
4
|
Wang K, Zhang T, Liu M, Wang D, Zhu H, Wang Z, Yu F, Liu Y, Zhao W. Synthesis and immunological evaluation of Mincle ligands-based antitumor vaccines. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.108065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
5
|
Khedri M, Afsharchi F, Souderjani AH, Rezvantalab S, Didandeh M, Maleki R, Musaie K, Santos HA, Shahbazi M. Molecular scale study on the interactions of biocompatible nanoparticles with macrophage membrane and blood proteins. NANO SELECT 2022. [DOI: 10.1002/nano.202200043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Mohammad Khedri
- Computational Biology and Chemistry Group (CBCG) Universal Scientific Education and Research Network (USERN) Tehran Iran
| | - Fatemeh Afsharchi
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC) Zanjan University of Medical Sciences Zanjan Iran
| | - Amirhosein Hasanpour Souderjani
- Department of Pharmaceutical Engineering, School of Chemical Engineering College of Engineering, University of Tehran Tehran Iran
| | - Sima Rezvantalab
- Renewable Energies Department Faculty of Chemical Engineering Urmia University of Technology Urmia Iran
| | - Mohsen Didandeh
- Department of Chemical Engineering Tarbiat Modares University Tehran Iran
| | - Reza Maleki
- Computational Biology and Chemistry Group (CBCG) Universal Scientific Education and Research Network (USERN) Tehran Iran
| | - Kiyan Musaie
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC) Zanjan University of Medical Sciences Zanjan Iran
| | - Hélder A. Santos
- Department of Biomedical Engineering University Medical Center Groningen University of Groningen Groningen The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science University of Groningen/University Medical Center Groningen Groningen The Netherlands
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki Finland
| | - Mohammad‐Ali Shahbazi
- Department of Biomedical Engineering University Medical Center Groningen University of Groningen Groningen The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science University of Groningen/University Medical Center Groningen Groningen The Netherlands
| |
Collapse
|
6
|
Braganza CD, Kodar K, Teunissen T, Andreassend SK, Khan A, Timmer MSM, Stocker BL. Lipophilic glucose monoesters and glycosides are potent human Mincle agonists. Org Biomol Chem 2022; 20:3096-3104. [DOI: 10.1039/d1ob02111g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Macrophage inducible C-type lectin (Mincle) is a pattern recognition receptor on myeloid cells that represents a promising target for Th1-stimulating adjuvants. We report on the synthesis of branched and aromatic...
Collapse
|
7
|
Neupane YR, Huang C, Wang X, Chng WH, Venkatesan G, Zharkova O, Wacker MG, Czarny B, Storm G, Wang JW, Pastorin G. Lyophilization Preserves the Intrinsic Cardioprotective Activity of Bioinspired Cell-Derived Nanovesicles. Pharmaceutics 2021; 13:pharmaceutics13071052. [PMID: 34371743 PMCID: PMC8309024 DOI: 10.3390/pharmaceutics13071052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022] Open
Abstract
Recently, bioinspired cell-derived nanovesicles (CDNs) have gained much interest in the field of nanomedicine due to the preservation of biomolecular structure characteristics derived from their parent cells, which impart CDNs with unique properties in terms of binding and uptake by target cells and intrinsic biological activities. Although the production of CDNs can be easily and reproducibly achieved with any kind of cell culture, application of CDNs for therapeutic purposes has been greatly hampered by their physical and chemical instability during long-term storage in aqueous dispersion. In the present study, we conceived a lyophilization approach that would preserve critical characteristics regarding stability (vesicles' size and protein content), structural integrity, and biological activity of CDNs for enabling long-term storage in freeze-dried form. Compared to the lyoprotectant sucrose, trehalose-lyoprotected CDNs showed significantly higher glass transition temperature and lower residual moisture content. As assessed by ATR-FTIR and far-UV circular dichroism, lyophilization in the presence of the lyoprotectant effectively maintained the secondary structure of cellular proteins. After reconstitution, lyoprotected CDNs were efficiently associated with HeLa cells, CT26 cells, and bone marrow-derived macrophages at a rate comparable to the freshly prepared CDNs. In vivo, both lyoprotected and freshly prepared CDNs, for the first time ever reported, targeted the injured heart, and exerted intrinsic cardioprotective effects within 24 h, attributable to the antioxidant capacity of CDNs in a myocardial ischemia/reperfusion injury animal model. Taken together, these results pave the way for further development of CDNs as cell-based therapeutics stabilized by lyophilization that enabled long-term storage while preserving their activity.
Collapse
Affiliation(s)
- Yub Raj Neupane
- Department of Pharmacy, National University of Singapore, Singapore 117559, Singapore; (Y.R.N.); (W.H.C.); (G.V.); (M.G.W.)
| | - Chenyuan Huang
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore; (C.H.); (X.W.); (O.Z.); (G.S.)
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore
| | - Xiaoyuan Wang
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore; (C.H.); (X.W.); (O.Z.); (G.S.)
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore
| | - Wei Heng Chng
- Department of Pharmacy, National University of Singapore, Singapore 117559, Singapore; (Y.R.N.); (W.H.C.); (G.V.); (M.G.W.)
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 119077, Singapore
| | - Gopalakrishnan Venkatesan
- Department of Pharmacy, National University of Singapore, Singapore 117559, Singapore; (Y.R.N.); (W.H.C.); (G.V.); (M.G.W.)
- Antimicrobial Resistance Interdisciplinary Research Group (AMR-IRG), Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Olga Zharkova
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore; (C.H.); (X.W.); (O.Z.); (G.S.)
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore
| | - Matthias Gerhard Wacker
- Department of Pharmacy, National University of Singapore, Singapore 117559, Singapore; (Y.R.N.); (W.H.C.); (G.V.); (M.G.W.)
| | - Bertrand Czarny
- School of Materials, Science and Engineering & Lee Kong Chian School of Medicine (LKC Medicine), Nanyang Technological University, Singapore 308232, Singapore;
| | - Gerrit Storm
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore; (C.H.); (X.W.); (O.Z.); (G.S.)
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, 3584 CS Utrecht, The Netherlands
- Department of Targeted Therapeutics, University of Twente, 7522 NB Enschede, The Netherlands
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117609, Singapore
| | - Jiong-Wei Wang
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore; (C.H.); (X.W.); (O.Z.); (G.S.)
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117609, Singapore
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore
- Correspondence: (J.-W.W.); (G.P.)
| | - Giorgia Pastorin
- Department of Pharmacy, National University of Singapore, Singapore 117559, Singapore; (Y.R.N.); (W.H.C.); (G.V.); (M.G.W.)
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 119077, Singapore
- NUSNNI-NanoCore, National University of Singapore, Singapore 117574, Singapore
- Correspondence: (J.-W.W.); (G.P.)
| |
Collapse
|
8
|
Socher E, Conrad M, Heger L, Paulsen F, Sticht H, Zunke F, Arnold P. Mutations in the B.1.1.7 SARS-CoV-2 Spike Protein Reduce Receptor-Binding Affinity and Induce a Flexible Link to the Fusion Peptide. Biomedicines 2021; 9:525. [PMID: 34066729 PMCID: PMC8151884 DOI: 10.3390/biomedicines9050525] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/05/2021] [Indexed: 02/08/2023] Open
Abstract
The B.1.1.7 variant of the SARS-CoV-2 virus shows enhanced infectiousness over the wild type virus, leading to increasing patient numbers in affected areas. Amino acid exchanges within the SARS-CoV-2 spike protein variant of B.1.1.7 affect inter-monomeric contact sites within the trimer (A570D and D614G) as well as the ACE2-receptor interface region (N501Y), which comprises the receptor-binding domain (RBD) of the spike protein. However, the molecular consequences of mutations within B.1.1.7 on spike protein dynamics and stability or ACE2 binding are largely unknown. Here, molecular dynamics simulations comparing SARS-CoV-2 wild type with the B.1.1.7 variant revealed inter-trimeric contact rearrangements, altering the structural flexibility within the spike protein trimer. Furthermore, we found increased flexibility in direct spatial proximity of the fusion peptide due to salt bridge rearrangements induced by the D614G mutation in B.1.1.7. This study also implies a reduced binding affinity for B.1.1.7 with ACE2, as the N501Y mutation restructures the RBD-ACE2 interface, significantly decreasing the linear interaction energy between the RBD and ACE2. Our results demonstrate how mutations found within B.1.1.7 enlarge the flexibility around the fusion peptide and change the RBD-ACE2 interface. We anticipate our findings to be starting points for in depth biochemical and cell biological analyses of B.1.1.7.
Collapse
Affiliation(s)
- Eileen Socher
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, 91054 Erlangen, Germany
| | - Marcus Conrad
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.C.); (H.S.)
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, 91052 Erlangen, Germany;
| | - Friedrich Paulsen
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
- Department of Operative Surgery and Topographic Anatomy, Sechenov University, 119992 Moscow, Russia
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (M.C.); (H.S.)
- Erlangen National High Performance Computing Center (NHR@FAU), Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), University Hospital Erlangen, 91054 Erlangen, Germany;
| | - Philipp Arnold
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
| |
Collapse
|
9
|
Miyake Y, Yamasaki S. Immune Recognition of Pathogen-Derived Glycolipids Through Mincle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1204:31-56. [DOI: 10.1007/978-981-15-1580-4_2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
10
|
Ryter KT, Ettenger G, Rasheed OK, Buhl C, Child R, Miller SM, Holley D, Smith AJ, Evans JT. Aryl Trehalose Derivatives as Vaccine Adjuvants for Mycobacterium tuberculosis. J Med Chem 2019; 63:309-320. [PMID: 31809053 DOI: 10.1021/acs.jmedchem.9b01598] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mycobacterium tuberculosis (Mtb) continues to be a major health threat worldwide, and the development of Mtb vaccines could play a pivotal role in the prevention and control of this devastating epidemic. Th17-mediated immunity has been implicated in disease protection correlates of immune protection against Mtb. Currently, there are no approved adjuvants capable of driving a Th17 response in a vaccine setting. Recent clinical trial results using trehalose dibehenate have demonstrated a formulation-dependant proof of concept adjuvant system CAF01 capable of inducing long-lived protection. We have discovered a new class of Th17-inducing vaccine adjuvants based on the natural product Brartemicin. We synthesized and evaluated the capacity of a library of aryl trehalose derivatives to drive immunostimulatory reresponses and evaluated the structure-activity relationships in terms of the ability to engage the Mincle receptor and induce production of innate cytokines from human and murine cells. We elaborated on the structure-activity relationship of the new scaffold and demonstrated the ability of the lead entity to induce a pro-Th17 cytokine profile from primary human peripheral blood mononuclear cells and demonstrated efficacy in generating antibodies in combination with tuberculosis antigen M72 in a mouse model.
Collapse
|
11
|
Reinink P, Buter J, Mishra VK, Ishikawa E, Cheng TY, Willemsen PTJ, Porwollik S, Brennan PJ, Heinz E, Mayfield JA, Dougan G, van Els CA, Cerundolo V, Napolitani G, Yamasaki S, Minnaard AJ, McClelland M, Moody DB, Van Rhijn I. Discovery of Salmonella trehalose phospholipids reveals functional convergence with mycobacteria. J Exp Med 2019; 216:757-771. [PMID: 30804000 PMCID: PMC6446866 DOI: 10.1084/jem.20181812] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 12/08/2018] [Accepted: 02/01/2019] [Indexed: 01/11/2023] Open
Abstract
Salmonella species are among the world's most prevalent pathogens. Because the cell wall interfaces with the host, we designed a lipidomics approach to reveal pathogen-specific cell wall compounds. Among the molecules differentially expressed between Salmonella Paratyphi and S. Typhi, we focused on lipids that are enriched in S. Typhi, because it causes typhoid fever. We discovered a previously unknown family of trehalose phospholipids, 6,6'-diphosphatidyltrehalose (diPT) and 6-phosphatidyltrehalose (PT). Cardiolipin synthase B (ClsB) is essential for PT and diPT but not for cardiolipin biosynthesis. Chemotyping outperformed clsB homology analysis in evaluating synthesis of diPT. DiPT is restricted to a subset of Gram-negative bacteria: large amounts are produced by S. Typhi, lower amounts by other pathogens, and variable amounts by Escherichia coli strains. DiPT activates Mincle, a macrophage activating receptor that also recognizes mycobacterial cord factor (6,6'-trehalose dimycolate). Thus, Gram-negative bacteria show convergent function with mycobacteria. Overall, we discovered a previously unknown immunostimulant that is selectively expressed among medically important bacterial species.
Collapse
Affiliation(s)
- Peter Reinink
- Department of Infectious Diseases and Immunology, School of Veterinary Medicine, Utrecht University, Utrecht, Netherlands.,Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Jeffrey Buter
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Vivek K Mishra
- Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
| | - Eri Ishikawa
- Department of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Tan-Yun Cheng
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Peter T J Willemsen
- Wageningen Bioveterinary Research, Department of Infection Biology, Lelystad, Netherlands
| | - Steffen Porwollik
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA
| | - Patrick J Brennan
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Eva Heinz
- Wellcome Trust Sanger Institute, Hinxton, UK
| | - Jacob A Mayfield
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA
| | | | - Cécile A van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Giorgio Napolitani
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Sho Yamasaki
- Department of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Adriaan J Minnaard
- Stratingh Institute for Chemistry, University of Groningen, Groningen, Netherlands
| | - Michael McClelland
- Department of Microbiology and Molecular Genetics, University of California, Irvine, Irvine, CA
| | - D Branch Moody
- Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA
| | - Ildiko Van Rhijn
- Department of Infectious Diseases and Immunology, School of Veterinary Medicine, Utrecht University, Utrecht, Netherlands .,Department of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital, Boston, MA
| |
Collapse
|
12
|
Söldner CA, Horn AHC, Sticht H. Binding of histamine to the H1 receptor-a molecular dynamics study. J Mol Model 2018; 24:346. [PMID: 30498974 DOI: 10.1007/s00894-018-3873-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 11/05/2018] [Indexed: 02/06/2023]
Abstract
Binding of histamine to the G-protein coupled histamine H1 receptor plays an important role in the context of allergic reactions; however, no crystal structure of the resulting complex is available yet. To deduce the histamine binding site, we performed unbiased molecular dynamics (MD) simulations on a microsecond time scale, which allowed to monitor one binding event, in which particularly the residues of the extracellular loop 2 were involved in the initial recognition process. The final histamine binding pose in the orthosteric pocket is characterized by interactions with Asp1073.32, Tyr1083.33, Thr1945.43, Asn1985.46, Trp4286.48, Tyr4316.51, Phe4326.52, and Phe4356.55, which is in agreement with existing mutational data. The conformational stability of the obtained complex structure was subsequently confirmed in 2 μs equilibrium MD simulations, and a metadynamics simulation proved that the detected binding site represents an energy minimum. A complementary investigation of a D107A mutant, which has experimentally been shown to abolish ligand binding, revealed that this exchange results in a significantly weaker interaction and enhanced ligand dynamics. This finding underlines the importance of the electrostatic interaction between the histamine ammonium group and the side chain of Asp1073.32 for histamine binding.
Collapse
Affiliation(s)
- Christian A Söldner
- Bioinformatik, Institut für Biochemie, Emil-Fischer-Centrum, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), , Fahrstraße 17, 91054, Erlangen, Germany
| | - Anselm H C Horn
- Bioinformatik, Institut für Biochemie, Emil-Fischer-Centrum, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), , Fahrstraße 17, 91054, Erlangen, Germany
| | - Heinrich Sticht
- Bioinformatik, Institut für Biochemie, Emil-Fischer-Centrum, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), , Fahrstraße 17, 91054, Erlangen, Germany.
| |
Collapse
|