1
|
Basaran R, Budhadev D, Kempf A, Nehlmeier I, Hondow N, Pöhlmann S, Guo Y, Zhou D. Probing scaffold size effects on multivalent lectin-glycan binding affinity, thermodynamics and antiviral properties using polyvalent glycan-gold nanoparticles. NANOSCALE 2024; 16:13962-13978. [PMID: 38984502 DOI: 10.1039/d4nr00484a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Multivalent lectin-glycan interactions (MLGIs) are pivotal for viral infections and immune regulation. Their structural and biophysical data are thus highly valuable, not only for understanding their basic mechanisms but also for designing potent glycoconjugate therapeutics against target MLGIs. However, such information for some important MGLIs remains poorly understood, greatly limiting research progress. We have recently developed densely glycosylated nanoparticles, e.g., ∼4 nm quantum dots (QDs) or ∼5 nm gold nanoparticles (GNPs), as mechanistic probes for MLGIs. Using two important model lectin viral receptors, DC-SIGN and DC-SIGNR, we have shown that these probes can not only offer sensitive fluorescence assays for quantifying MLGI affinities, but also reveal key structural information (e.g., binding site orientation and binding mode) useful for MLGI targeting. However, the small sizes of the previous scaffolds may not be optimal for maximising MLGI affinity and targeting specificity. Herein, using α-manno-α-1,2-biose (DiMan) functionalised GNP (GNP-DiMan) probes, we have systematically studied how GNP scaffold size (e.g., 5, 13, and 27 nm) and glycan density (e.g., 100, 75, 50 and 25%) determine their MLGI affinities, thermodynamics, and antiviral properties. We have developed a new GNP fluorescence quenching assay format to minimise the possible interference of GNP's strong inner filter effect in MLGI affinity quantification, revealing that increasing the GNP size is highly beneficial for enhancing MLGI affinity. We have further determined the MLGI thermodynamics by combining temperature-dependent affinity and Van't Hoff analyses, revealing that GNP-DiMan-DC-SIGN/R binding is enthalpy driven with favourable binding Gibbs free energy changes (ΔG°) being enhanced with increasing GNP size. Finally, we show that increasing the GNP size significantly enhances their antiviral potency. Notably, the DiMan coated 27 nm GNP potently and robustly blocks both DC-SIGN and DC-SIGNR mediated pseudo-Ebola virus cellular entry with an EC50 of ∼23 and ∼49 pM, respectively, making it the most potent glycoconjugate inhibitor against DC-SIGN/R-mediated Ebola cellular infections. Our results have established GNP-glycans as a new tool for quantifying MLGI biophysical parameters and revealed that increasing the GNP scaffold size significantly enhances their MLGI affinities and antiviral potencies.
Collapse
Affiliation(s)
- Rahman Basaran
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Darshita Budhadev
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| | - Amy Kempf
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Nicole Hondow
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, UK
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Centre - Leibniz Institute for Primate Research, 37077 Göttingen, Germany
- Faculty of Biology and Psychology, Georg-August-University Göttingen, 37073 Göttingen, Germany
| | - Yuan Guo
- School of Food Science and Nutrition, and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Dejian Zhou
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
2
|
Li ZZ, Zhong NN, Cao LM, Cai ZM, Xiao Y, Wang GR, Liu B, Xu C, Bu LL. Nanoparticles Targeting Lymph Nodes for Cancer Immunotherapy: Strategies and Influencing Factors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308731. [PMID: 38327169 DOI: 10.1002/smll.202308731] [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: 09/29/2023] [Revised: 01/07/2024] [Indexed: 02/09/2024]
Abstract
Immunotherapy has emerged as a potent strategy in cancer treatment, with many approved drugs and modalities in the development stages. Despite its promise, immunotherapy is not without its limitations, including side effects and suboptimal efficacy. Using nanoparticles (NPs) as delivery vehicles to target immunotherapy to lymph nodes (LNs) can improve the efficacy of immunotherapy drugs and reduce side effects in patients. In this context, this paper reviews the development of LN-targeted immunotherapeutic NP strategies, the mechanisms of NP transport during LN targeting, and their related biosafety risks. NP targeting of LNs involves either passive targeting, influenced by NP physical properties, or active targeting, facilitated by affinity ligands on NP surfaces, while alternative methods, such as intranodal injection and high endothelial venule (HEV) targeting, have uncertain clinical applicability and require further research and validation. LN targeting of NPs for immunotherapy can reduce side effects and increase biocompatibility, but risks such as toxicity, organ accumulation, and oxidative stress remain, although strategies such as biodegradable biomacromolecules, polyethylene glycol (PEG) coating, and impurity addition can mitigate these risks. Additionally, this work concludes with a future-oriented discussion, offering critical insights into the field.
Collapse
Affiliation(s)
- Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Ze-Min Cai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| | - Chun Xu
- School of Dentistry, The University of Queensland, 288 Herston Road, Brisbane, 4066, Australia
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, #237 Luoyu Road, Wuhan, 430079, China
| |
Collapse
|
3
|
Basaran R, Ning X, Budhadev D, Hondow N, Guo Y, Zhou D. Probing the pH-dependency of DC-SIGN/R multivalent lectin-glycan interactions using polyvalent glycan-gold nanoparticles. NANOSCALE ADVANCES 2024; 6:2198-2208. [PMID: 38633047 PMCID: PMC11019501 DOI: 10.1039/d3na01013a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024]
Abstract
The dendritic cell tetrameric lectin, DC-SIGN, and its closely related endothelial cell lectin, DC-SIGNR (collectively abbreviated as DC-SIGN/R) play a key role in the binding and transmission of deadly viruses, including Ebola, HIV, HCV, and SARS-CoV-2. Their virus binding/release processes involve a gradually acidifying environment following the natural intracellular trafficking pathways. Therefore, understanding DC-SIGN/R's pH-dependent binding properties with glycan ligands is of great importance. We have recently developed densely glycosylated gold nanoparticles (glycan-GNPs) as a powerful new tool for probing DC-SIGN/R multivalent lectin-glycan interaction (MLGI) mechanisms. They can provide not only quantitative MLGI affinities but also important structural information, such as binding site orientation and binding modes. Herein, we further employ the glycan-GNP probes to investigate the pH dependency of DC-SIGN/R MLGI properties. We find that DC-SIGN/R MLGIs exhibit distinct pH dependence over the normal physiological (7.4) to lysosomal (∼4.6) pH range. DC-SIGN binds glycan-GNPs strongly and stably from pH 7.4 to ∼5.8, but the binding is weakened significantly as pH decreases to ≤5.4 and may be fully dissociated at pH 4.6. This behaviour is fully consistent with DC-SIGN's role as an endocytic recycling receptor. In contrast, DC-SIGNR's affinity with glycan-GNPs is enhanced with the decreasing pH from 7.4 to 5.4, peaking at pH 5.4, and then reduced as pH is further lowered. Interestingly, both DC-SIGN/R binding with glycan-GNPs are found to be partially reversible in a pH-dependent manner.
Collapse
Affiliation(s)
- Rahman Basaran
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Xinyu Ning
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Darshita Budhadev
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Nicole Hondow
- School of Chemical and Process Engineering, University of Leeds Leeds LS2 9JT UK
| | - Yuan Guo
- School of Food Science and Nutrition, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Dejian Zhou
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| |
Collapse
|
4
|
Bains A, Fischer K, Guan W, LiWang PJ. The Antiviral Activity of the Lectin Griffithsin against SARS-CoV-2 Is Enhanced by the Presence of Structural Proteins. Viruses 2023; 15:2452. [PMID: 38140693 PMCID: PMC10747160 DOI: 10.3390/v15122452] [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: 10/14/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Although COVID-19 transmission has been reduced by the advent of vaccinations and a variety of rapid monitoring techniques, the SARS-CoV-2 virus itself has shown a remarkable ability to mutate and persist. With this long track record of immune escape, researchers are still exploring prophylactic treatments to curtail future SARS-CoV-2 variants. Specifically, much focus has been placed on the antiviral lectin Griffithsin in preventing spike protein-mediated infection via the hACE2 receptor (direct infection). However, an oft-overlooked aspect of SARS-CoV-2 infection is viral capture by attachment receptors such as DC-SIGN, which is thought to facilitate the initial stages of COVID-19 infection in the lung tissue (called trans-infection). In addition, while immune escape is dictated by mutations in the spike protein, coronaviral virions also incorporate M, N, and E structural proteins within the particle. In this paper, we explored how several structural facets of both the SARS-CoV-2 virion and the antiviral lectin Griffithsin can affect and attenuate the infectivity of SARS-CoV-2 pseudovirus. We found that Griffithsin was a better inhibitor of hACE2-mediated direct infection when the coronaviral M protein is present compared to when it is absent (possibly providing an explanation regarding why Griffithsin shows better inhibition against authentic SARS-CoV-2 as opposed to pseudotyped viruses, which generally do not contain M) and that Griffithsin was not an effective inhibitor of DC-SIGN-mediated trans-infection. Furthermore, we found that DC-SIGN appeared to mediate trans-infection exclusively via binding to the SARS-CoV-2 spike protein, with no significant effect observed when other viral proteins (M, N, and/or E) were present. These results provide etiological data that may help to direct the development of novel antiviral treatments, either by leveraging Griffithsin binding to the M protein as a novel strategy to prevent SARS-CoV-2 infection or by narrowing efforts to inhibit trans-infection to focus on DC-SIGN binding to SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
- Arjan Bains
- Chemistry and Biochemistry, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Kathryn Fischer
- Quantitative and Systems Biology, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Wenyan Guan
- Materials and Biomaterials Science and Engineering, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Patricia J. LiWang
- Molecular Cell Biology, Health Sciences Research Institute, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA
| |
Collapse
|
5
|
Martínez-Bailén M, Rojo J, Ramos-Soriano J. Multivalent glycosystems for human lectins. Chem Soc Rev 2023; 52:536-572. [PMID: 36545903 DOI: 10.1039/d2cs00736c] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Human lectins are involved in a wide variety of biological processes, both physiological and pathological, which have attracted the interest of the scientific community working in the glycoscience field. Multivalent glycosystems have been employed as useful tools to understand carbohydrate-lectin binding processes as well as for biomedical applications. The review shows the different scaffolds designed for a multivalent presentation of sugars and their corresponding binding studies to lectins and in some cases, their biological activities. We summarise this research by organizing based on lectin types to highlight the progression in this active field. The paper provides an overall picture of how these contributions have furnished relevant information on this topic to help in understanding and participate in these carbohydrate-lectin interactions.
Collapse
Affiliation(s)
- Macarena Martínez-Bailén
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Av. Américo Vespucio 49, Seville 41092, Spain.
| | - Javier Rojo
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Av. Américo Vespucio 49, Seville 41092, Spain.
| | - Javier Ramos-Soriano
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Av. Américo Vespucio 49, Seville 41092, Spain.
| |
Collapse
|
6
|
Hooper J, Liu Y, Budhadev D, Ainaga DF, Hondow N, Zhou D, Guo Y. Polyvalent Glycan Quantum Dots as a Multifunctional Tool for Revealing Thermodynamic, Kinetic, and Structural Details of Multivalent Lectin-Glycan Interactions. ACS APPLIED MATERIALS & INTERFACES 2022; 14:47385-47396. [PMID: 36194567 PMCID: PMC9614721 DOI: 10.1021/acsami.2c11111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Multivalent lectin-glycan interactions (MLGIs) are widespread and vital for biology. Their binding biophysical and structural details are thus highly valuable, not only for the understanding of binding affinity and specificity mechanisms but also for guiding the design of multivalent therapeutics against specific MLGIs. However, effective techniques that can reveal all such details remain unavailable. We have recently developed polyvalent glycan quantum dots (glycan-QDs) as a new probe for MLGIs. Using a pair of closely related tetrameric viral-binding lectins, DC-SIGN and DC-SIGNR, as model examples, we have revealed and quantified their large affinity differences in glycan-QD binding are due to distinct binding modes: with simultaneous binding for DC-SIGN and cross-linking for DC-SIGNR. Herein, we further extend the capacity of the glycan-QD probes by investigating the correlation between binding mode and binding thermodynamics and kinetics and further probing a structural basis of their binding nature. We reveal that while both lectins' binding with glycan-QDs is enthalpy driven with similar binding enthalpy changes, DC-SIGN pays a lower binding entropy penalty, resulting in a higher affinity than DC-SIGNR. We then show that DC-SIGN binding gives a single second-order kon rate, whereas DC-SIGNR gives a rapid initial binding followed by a much slower secondary interaction. We further identify a structural element in DC-SIGN, absent in DC-SIGNR, that plays an important role in maintaining DC-SIGN's MLGI character. Its removal switches the binding from being enthalpically to entropically driven and gives mixed binding modes containing both simultaneous and cross-linking binding behavior, without markedly affecting the overall binding affinity and kinetics.
Collapse
Affiliation(s)
- James Hooper
- School
of Food Science & Nutrition and Astbury Centre for Structural
Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Yuanyuan Liu
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United
Kingdom
| | - Darshita Budhadev
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United
Kingdom
| | - Dario Fernandez Ainaga
- School
of Chemical and Process Engineering, University
of Leeds, Leeds LS2 9JT, United Kingdom
| | - Nicole Hondow
- School
of Chemical and Process Engineering, University
of Leeds, Leeds LS2 9JT, United Kingdom
| | - Dejian Zhou
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United
Kingdom
| | - Yuan Guo
- School
of Food Science & Nutrition and Astbury Centre for Structural
Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
7
|
Pappalardo JS, Salmaso S, Levchenko TS, Mastrotto F, Bersani S, Langellotti CA, Vermeulen M, Ghersa F, Quattrocchi V, Zamorano PI, Hartner WC, Toniutti M, Musacchio T, Torchilin VP. Characterization of a Nanovaccine Platform Based on an α1,2-Mannobiose Derivative Shows Species-non-specific Targeting to Human, Bovine, Mouse, and Teleost Fish Dendritic Cells. Mol Pharm 2021; 18:2540-2555. [PMID: 34106726 DOI: 10.1021/acs.molpharmaceut.1c00048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Dendritic cells serve as the main immune cells that trigger the immune response. We developed a simple and cost-effective nanovaccine platform based on the α1',2-mannobiose derivative for dendritic cell targeting. In previous work, we have formulated the α1,2-mannobiose-based nanovaccine platform with plasmid DNA and tested it in cattle against BoHV-1 infection. There, we have shown that the dendritic cell targeting using this nanovaccine platform in vivo can boost the immunogenicity, resulting in a long-lasting immunity. In this work, we aim to characterize the α1',2-mannobiose derivative, which is key in the nanovaccine platform. This DC-targeting strategy takes advantage of the specific receptor known as DC-SIGN and exploits its capacity to bind α1,2-mannobiose that is present at terminal ends of oligosaccharides in certain viruses, bacteria, and other pathogens. The oxidative conjugation of α1',2-mannobiose to NH2-PEG2kDa-DSPE allowed us to preserve the chemical structure of the non-reducing mannose of the disaccharide and the OH groups and the stereochemistry of all carbons of the reducing mannose involved in the binding to DC-SIGN. Here, we show specific targeting to DC-SIGN of decorated micelles incubated with the Raji/DC-SIGN cell line and uptake of targeted liposomes that took place in human, bovine, mouse, and teleost fish DCs in vitro, by flow cytometry. Specific targeting was found in all cultures, demonstrating a species-non-specific avidity for this ligand, which opens up the possibility of using this nanoplatform to develop new vaccines for various species, including humans.
Collapse
Affiliation(s)
- Juan Sebastian Pappalardo
- Veterinary Nanomedicine Group, Instituto de Investigaciones Forestales y Agropecuarias Bariloche (IFAB, INTA-CONICET), EEA Bariloche, Instituto Nacional de Tecnología Agropecuaria, Bote Modesta Victoria 4450, San Carlos de Bariloche, Río Negro R8403DVZ, Argentina.,Immunology and Immunomodulators Group, Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), IV, Instituto Nacional de Tecnología Agropecuaria, Nicolás Repetto 2799, William Morris, Buenos Aires B1681FUU, Argentina.,Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padova, Via F. Marzolo, 5, Padova 35121, Padova, Italy
| | - Tatyana S Levchenko
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padova, Via F. Marzolo, 5, Padova 35121, Padova, Italy
| | - Sara Bersani
- Department of Pharmaceutical and Pharmacological Sciences, School of Medicine, University of Padova, Via F. Marzolo, 5, Padova 35121, Padova, Italy
| | - Cecilia A Langellotti
- Immunology and Immunomodulators Group, Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), IV, Instituto Nacional de Tecnología Agropecuaria, Nicolás Repetto 2799, William Morris, Buenos Aires B1681FUU, Argentina.,National Council of Scientific and Technical Research (CONICET), Avenida Rivadavia 1917, Ciudad de Buenos Aires C1033AAJ, Argentina
| | - Monica Vermeulen
- National Council of Scientific and Technical Research (CONICET), Avenida Rivadavia 1917, Ciudad de Buenos Aires C1033AAJ, Argentina.,Institute of Experimental Medicine (IMEX, ANM-CONICET), Academia Nacional de Medicina, Pacheco de Melo 3081, Ciudad de Buenos Aires C1425AUM, Argentina
| | - Federica Ghersa
- Veterinary Nanomedicine Group, Instituto de Investigaciones Forestales y Agropecuarias Bariloche (IFAB, INTA-CONICET), EEA Bariloche, Instituto Nacional de Tecnología Agropecuaria, Bote Modesta Victoria 4450, San Carlos de Bariloche, Río Negro R8403DVZ, Argentina.,Parasitology Laboratory, Instituto de Investigaciones en Biodiversidad y Medioambiente (INIBIOMA, UNCo-CONICET) Universidad Nacional del Comahue, Quintral 1250, San Carlos de Bariloche, Río Negro R8400FRF, Argentina
| | - Valeria Quattrocchi
- Immunology and Immunomodulators Group, Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), IV, Instituto Nacional de Tecnología Agropecuaria, Nicolás Repetto 2799, William Morris, Buenos Aires B1681FUU, Argentina
| | - Patricia I Zamorano
- Immunology and Immunomodulators Group, Instituto de Virología e Innovaciones Tecnológicas (IVIT, INTA-CONICET), IV, Instituto Nacional de Tecnología Agropecuaria, Nicolás Repetto 2799, William Morris, Buenos Aires B1681FUU, Argentina.,National Council of Scientific and Technical Research (CONICET), Avenida Rivadavia 1917, Ciudad de Buenos Aires C1033AAJ, Argentina
| | - William C Hartner
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Micaela Toniutti
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Tiziana Musacchio
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
8
|
Budhadev D, Poole E, Nehlmeier I, Liu Y, Hooper J, Kalverda E, Akshath US, Hondow N, Turnbull WB, Pöhlmann S, Guo Y, Zhou D. Glycan-Gold Nanoparticles as Multifunctional Probes for Multivalent Lectin-Carbohydrate Binding: Implications for Blocking Virus Infection and Nanoparticle Assembly. J Am Chem Soc 2020; 142:18022-18034. [PMID: 32935985 DOI: 10.1021/jacs.0c06793] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multivalent lectin-glycan interactions are widespread in biology and are often exploited by pathogens to bind and infect host cells. Glycoconjugates can block such interactions and thereby prevent infection. The inhibition potency strongly depends on matching the spatial arrangement between the multivalent binding partners. However, the structural details of some key lectins remain unknown and different lectins may exhibit overlapping glycan specificity. This makes it difficult to design a glycoconjugate that can potently and specifically target a particular multimeric lectin for therapeutic interventions, especially under the challenging in vivo conditions. Conventional techniques such as surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC) can provide quantitative binding thermodynamics and kinetics. However, they cannot reveal key structural information, e.g., lectin's binding site orientation, binding mode, and interbinding site spacing, which are critical to design specific multivalent inhibitors. Herein we report that gold nanoparticles (GNPs) displaying a dense layer of simple glycans are powerful mechanistic probes for multivalent lectin-glycan interactions. They can not only quantify the GNP-glycan-lectin binding affinities via a new fluorescence quenching method, but also reveal drastically different affinity enhancing mechanisms between two closely related tetrameric lectins, DC-SIGN (simultaneous binding to one GNP) and DC-SIGNR (intercross-linking with multiple GNPs), via a combined hydrodynamic size and electron microscopy analysis. Moreover, a new term, potential of assembly formation (PAF), has been proposed to successfully predict the assembly outcomes based on the binding mode between GNP-glycans and lectins. Finally, the GNP-glycans can potently and completely inhibit DC-SIGN-mediated augmentation of Ebola virus glycoprotein-driven cell entry (with IC50 values down to 95 pM), but only partially block DC-SIGNR-mediated virus infection. Our results suggest that the ability of a glycoconjugate to simultaneously block all binding sites of a target lectin is key to robust inhibition of viral infection.
Collapse
Affiliation(s)
- Darshita Budhadev
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Emma Poole
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research and Faculty of Biology and Psychology, University of Göttingen, Göttingen 37073, Germany
| | - Yuanyuan Liu
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - James Hooper
- School of Food Science & Nutrition and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Elizabeth Kalverda
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Uchangi Satyaprasad Akshath
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Nicole Hondow
- School of Chemical and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center-Leibniz Institute for Primate Research and Faculty of Biology and Psychology, University of Göttingen, Göttingen 37073, Germany
| | - Yuan Guo
- School of Food Science & Nutrition and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Dejian Zhou
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
9
|
Abstract
Multivalent protein-protein interactions serve central roles in many essential biological processes, ranging from cell signaling and adhesion to pathogen recognition. Uncovering the rules that govern these intricate interactions is important not only to basic biology and chemistry but also to the applied sciences where researchers are interested in developing molecules to promote or inhibit these interactions. Here we report the synthesis and application of atomically precise inorganic cluster nanomolecules consisting of an inorganic core and a covalently linked densely packed layer of saccharides. These hybrid agents are stable under biologically relevant conditions and exhibit multivalent binding capabilities, which enable us to study the complex interactions between glycosylated structures and a dendritic cell lectin receptor. Importantly, we find that subtle changes in the molecular structure lead to significant differences in the nanomolecule's protein-binding properties. Furthermore, we demonstrate an example of using these hybrid nanomolecules to effectively inhibit protein-protein interactions in a human cell line. Ultimately, this work reveals an intricate interplay between the structural design of multivalent agents and their biological activities toward protein surfaces.
Collapse
|
10
|
Tirumuru N, Wu L. HIV-1 envelope proteins up-regulate N6-methyladenosine levels of cellular RNA independently of viral replication. J Biol Chem 2019; 294:3249-3260. [PMID: 30617182 PMCID: PMC6398121 DOI: 10.1074/jbc.ra118.005608] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/04/2019] [Indexed: 01/09/2023] Open
Abstract
N6-methyladenosine (m6A) modification of HIV-1 RNA regulates viral replication and protein expression. The m6A modification is regulated by two groups of cellular proteins named writers and erasers that add or remove m6A, respectively. HIV-1 infection of CD4+ T-cells increases m6A levels of cellular mRNA, but the underlying mechanism is unknown. Here, we show that HIV-1 infection of CD4+ primary T-cells or Jurkat cells significantly increases m6A levels of cellular RNA independently of viral replication. Compared with HIV-1-infected CD4+ T-cells, similar m6A up-regulation was detected in total RNA from HIV-1-infected cells treated with a reverse-transcriptase inhibitor or with heat-inactivated HIV-1. Compared with mock controls, significantly increased m6A levels were detected in total RNA from Jurkat cells infected by single-cycle HIV-1 pseudotyped with an HIV-1 envelope (Env) glycoprotein, but not with vesicular stomatitis virus glycoprotein G (VSV-G). Overexpression of HIV-1 Env in HEK293T cells did not affect m6A levels of cellular RNA, suggesting that de novo synthesis of Env is not required for m6A up-regulation. Interestingly, treatment of Jurkat cells with recombinant gp120 of HIV-1 Env significantly increased m6A levels of cellular RNA, which was reduced by a gp120-neutralizing antibody. Preincubation of Jurkat cells with a CD4 receptor-neutralizing antibody blocked HIV-1-induced up-regulation of m6A levels in cellular RNA. Moreover, HIV-1 infection or gp120 treatment did not alter the protein expression of m6A writers and erasers in cells. Our findings suggest that HIV-1 gp120 binding to the CD4 receptor is required for m6A up-regulation in cells.
Collapse
Affiliation(s)
- Nagaraja Tirumuru
- From the Center for Retrovirus Research, Department of Veterinary Biosciences and
- the Center for RNA Biology, Ohio State University, Columbus, Ohio 43210
| | - Li Wu
- From the Center for Retrovirus Research, Department of Veterinary Biosciences and
- the Center for RNA Biology, Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
11
|
Probing Multivalent Protein–Carbohydrate Interactions by Quantum Dot-Förster Resonance Energy Transfer. Methods Enzymol 2018; 598:71-100. [DOI: 10.1016/bs.mie.2017.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
12
|
Guo Y, Nehlmeier I, Poole E, Sakonsinsiri C, Hondow N, Brown A, Li Q, Li S, Whitworth J, Li Z, Yu A, Brydson R, Turnbull WB, Pöhlmann S, Zhou D. Dissecting Multivalent Lectin-Carbohydrate Recognition Using Polyvalent Multifunctional Glycan-Quantum Dots. J Am Chem Soc 2017; 139:11833-11844. [PMID: 28786666 PMCID: PMC5579584 DOI: 10.1021/jacs.7b05104] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Indexed: 01/05/2023]
Abstract
Multivalent protein-carbohydrate interactions initiate the first contacts between virus/bacteria and target cells, which ultimately lead to infection. Understanding the structures and binding modes involved is vital to the design of specific, potent multivalent inhibitors. However, the lack of structural information on such flexible, complex, and multimeric cell surface membrane proteins has often hampered such endeavors. Herein, we report that quantum dots (QDs) displayed with a dense array of mono-/disaccharides are powerful probes for multivalent protein-glycan interactions. Using a pair of closely related tetrameric lectins, DC-SIGN and DC-SIGNR, which bind to the HIV and Ebola virus glycoproteins (EBOV-GP) to augment viral entry and infect target cells, we show that such QDs efficiently dissect the different DC-SIGN/R-glycan binding modes (tetra-/di-/monovalent) through a combination of multimodal readouts: Förster resonance energy transfer (FRET), hydrodynamic size measurement, and transmission electron microscopy imaging. We also report a new QD-FRET method for quantifying QD-DC-SIGN/R binding affinity, revealing that DC-SIGN binds to the QD >100-fold tighter than does DC-SIGNR. This result is consistent with DC-SIGN's higher trans-infection efficiency of some HIV strains over DC-SIGNR. Finally, we show that the QDs potently inhibit DC-SIGN-mediated enhancement of EBOV-GP-driven transduction of target cells with IC50 values down to 0.7 nM, matching well to their DC-SIGN binding constant (apparent Kd = 0.6 nM) measured by FRET. These results suggest that the glycan-QDs are powerful multifunctional probes for dissecting multivalent protein-ligand recognition and predicting glyconanoparticle inhibition of virus infection at the cellular level.
Collapse
Affiliation(s)
- Yuan Guo
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Inga Nehlmeier
- Infection
Biology Unit, German Primate Center, Kellnerweg 4, Gottingen 37077, Germany
| | - Emma Poole
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Chadamas Sakonsinsiri
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Nicole Hondow
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Andy Brown
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Qing Li
- Department
of Chemical Biology, Peking University Health
Sciences Centre, Beijing 100191, People’s Republic
of China
| | - Shuang Li
- Department
of Chemistry, Renmin University of China, Beijing 100872, People’s Republic of China
| | - Jessie Whitworth
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Zhongjun Li
- Department
of Chemical Biology, Peking University Health
Sciences Centre, Beijing 100191, People’s Republic
of China
| | - Anchi Yu
- Department
of Chemistry, Renmin University of China, Beijing 100872, People’s Republic of China
| | - Rik Brydson
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - W. Bruce Turnbull
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Stefan Pöhlmann
- Infection
Biology Unit, German Primate Center, Kellnerweg 4, Gottingen 37077, Germany
| | - Dejian Zhou
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, and School of Chemical
and Process Engineering, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
13
|
Dos Santos Á, Hadjivasiliou A, Ossa F, Lim NK, Turgut A, Taylor ME, Drickamer K. Oligomerization domains in the glycan-binding receptors DC-SIGN and DC-SIGNR: Sequence variation and stability differences. Protein Sci 2016; 26:306-316. [PMID: 27859859 PMCID: PMC5275740 DOI: 10.1002/pro.3083] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/21/2016] [Accepted: 11/08/2016] [Indexed: 11/05/2022]
Abstract
Human dendritic cell-specific intercellular adhesion molecule-1 grabbing nonintegrin, DC-SIGN, and the sinusoidal endothelial cell receptor DC-SIGNR or L-SIGN, are closely related sugar-binding receptors. DC-SIGN acts both as a pathogen-binding endocytic receptor and as a cell adhesion molecule, while DC-SIGNR has only the pathogen-binding function. In addition to differences in the sugar-binding properties of the carbohydrate-recognition domains in the two receptors, there are sequence differences in the adjacent neck domains, which are coiled-coil tetramerization domains comprised largely of 23-amino acid repeat units. A series of model polypeptides consisting of uniform repeat units have been characterized by gel filtration, differential scanning calorimetry and circular dichroism. The results demonstrate that two features characterize repeat units which form more stable tetramers: a leucine reside in the first position of the heptad pattern of hydrophobic residues that pack on the inside of the coiled coil and an arginine residue on the surface of the coiled coil that forms a salt bridge with a glutamic acid residue in the same polypeptide chain. In DC-SIGNR from all primates, very stable repeat units predominate, so the carbohydrate-recognition domains must be held relatively closely together. In contrast, stable repeat units are found only near the membrane in DC-SIGN. The presence of residues that disrupt tetramer formation in repeat units near the carbohydrate-recognition domains of DC-SIGN would allow these domains to splay further apart. Thus, the neck domains of DC-SIGN and DC-SIGNR can contribute to the different functions of these receptors by presenting the sugar-binding sites in different contexts.
Collapse
Affiliation(s)
- Ália Dos Santos
- Department of Life Sciences, Imperial College, London, SW7 2AZ, United Kingdom
| | | | - Felipe Ossa
- Department of Life Sciences, Imperial College, London, SW7 2AZ, United Kingdom
| | - Novandy K Lim
- Department of Life Sciences, Imperial College, London, SW7 2AZ, United Kingdom
| | - Aylin Turgut
- Department of Life Sciences, Imperial College, London, SW7 2AZ, United Kingdom
| | - Maureen E Taylor
- Department of Life Sciences, Imperial College, London, SW7 2AZ, United Kingdom
| | - Kurt Drickamer
- Department of Life Sciences, Imperial College, London, SW7 2AZ, United Kingdom
| |
Collapse
|
14
|
Guo Y, Sakonsinsiri C, Nehlmeier I, Fascione MA, Zhang H, Wang W, Pöhlmann S, Turnbull WB, Zhou D. Compact, Polyvalent Mannose Quantum Dots as Sensitive, Ratiometric FRET Probes for Multivalent Protein-Ligand Interactions. Angew Chem Int Ed Engl 2016; 55:4738-42. [PMID: 26990806 PMCID: PMC4979658 DOI: 10.1002/anie.201600593] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Indexed: 12/22/2022]
Abstract
A highly efficient cap-exchange approach for preparing compact, dense polyvalent mannose-capped quantum dots (QDs) has been developed. The resulting QDs have been successfully used to probe multivalent interactions of HIV/Ebola receptors DC-SIGN and DC-SIGNR (collectively termed as DC-SIGN/R) using a sensitive, ratiometric Förster resonance energy transfer (FRET) assay. The QD probes specifically bind DC-SIGN, but not its closely related receptor DC-SIGNR, which is further confirmed by its specific blocking of DC-SIGN engagement with the Ebola virus glycoprotein. Tuning the QD surface mannose valency reveals that DC-SIGN binds more efficiently to densely packed mannosides. A FRET-based thermodynamic study reveals that the binding is enthalpy-driven. This work establishes QD FRET as a rapid, sensitive technique for probing structure and thermodynamics of multivalent protein-ligand interactions.
Collapse
Affiliation(s)
- Yuan Guo
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| | - Chadamas Sakonsinsiri
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Inga Nehlmeier
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077, Göttingen, Germany
| | - Martin A Fascione
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Haiyan Zhang
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Weili Wang
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Kellnerweg 4, 37077, Göttingen, Germany
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Dejian Zhou
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
15
|
Guo Y, Sakonsinsiri C, Nehlmeier I, Fascione MA, Zhang H, Wang W, Pöhlmann S, Turnbull WB, Zhou D. Compact, Polyvalent Mannose Quantum Dots as Sensitive, Ratiometric FRET Probes for Multivalent Protein-Ligand Interactions. ACTA ACUST UNITED AC 2016; 128:4816-4820. [PMID: 27563159 PMCID: PMC4979676 DOI: 10.1002/ange.201600593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Indexed: 12/24/2022]
Abstract
A highly efficient cap-exchange approach for preparing compact, dense polyvalent mannose-capped quantum dots (QDs) has been developed. The resulting QDs have been successfully used to probe multivalent interactions of HIV/Ebola receptors DC-SIGN and DC-SIGNR (collectively termed as DC-SIGN/R) using a sensitive, ratiometric Förster resonance energy transfer (FRET) assay. The QD probes specifically bind DC-SIGN, but not its closely related receptor DC-SIGNR, which is further confirmed by its specific blocking of DC-SIGN engagement with the Ebola virus glycoprotein. Tuning the QD surface mannose valency reveals that DC-SIGN binds more efficiently to densely packed mannosides. A FRET-based thermodynamic study reveals that the binding is enthalpy-driven. This work establishes QD FRET as a rapid, sensitive technique for probing structure and thermodynamics of multivalent protein-ligand interactions.
Collapse
Affiliation(s)
- Yuan Guo
- School of Chemistry and Astbury Centre for Structural Molecular Biology University of Leeds Leeds LS2 9JT UK
| | - Chadamas Sakonsinsiri
- School of Chemistry and Astbury Centre for Structural Molecular Biology University of Leeds Leeds LS2 9JT UK
| | - Inga Nehlmeier
- Infection Biology Unit German Primate Center Kellnerweg 4 37077 Göttingen Germany
| | - Martin A Fascione
- Department of Chemistry University of York Heslington York YO10 5DD UK
| | - Haiyan Zhang
- School of Chemistry and Astbury Centre for Structural Molecular Biology University of Leeds Leeds LS2 9JT UK
| | - Weili Wang
- School of Chemistry and Astbury Centre for Structural Molecular Biology University of Leeds Leeds LS2 9JT UK
| | - Stefan Pöhlmann
- Infection Biology Unit German Primate Center Kellnerweg 4 37077 Göttingen Germany
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology University of Leeds Leeds LS2 9JT UK
| | - Dejian Zhou
- School of Chemistry and Astbury Centre for Structural Molecular Biology University of Leeds Leeds LS2 9JT UK
| |
Collapse
|
16
|
Human Blood-Circulating Basophils Capture HIV-1 and Mediate Viral trans-Infection of CD4+ T Cells. J Virol 2015; 89:8050-62. [PMID: 26018157 DOI: 10.1128/jvi.01021-15] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 05/18/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Cell-associated HIV-1 infection has been proposed to play a pivotal role in the spread of HIV-1 infection. Granulocytes are a category of white blood cells, comprising mainly basophils, neutrophils, and eosinophils, and participate in various inflammatory reactions and defense against pathogens. Here, we investigated the role of human blood granulocytes in the dissemination of HIV-1. These cells were found to express a variety of HIV-1 attachment factors (HAFs). Basophils expressed HAFs dendritic cell (DC)-specific intercellular adhesion molecule 3 (ICAM3)-grabbing nonintegrin (DC-SIGN), DC immunoreceptor (DCIR), heparan sulfate proteoglycan (HSPG), and α4β7 integrin and mediated the most efficient capture of HIV-1 on the cell surface. Neutrophils were found to express DCIR and demonstrated limited efficiency of viral capture. Eosinophils expressed α4β7 integrin but exhibited little or no virus-binding capacity. Intriguingly, following direct contact with CD4+ T cells, viruses harbored on the surface of basophils were transferred to T cells. The contact between basophils and CD4+ T cells and formation of infectious synapses appeared necessary for efficient HIV-1 spread. In HIV-1-infected individuals, the frequency of basophils remained fairly stable over the course of disease, regardless of CD4+ T depletion or the emergence of AIDS-associated opportunistic infections. Collectively, our results provide novel insights into the roles of granulocytes, particularly basophils, in HIV-1 dissemination. Thus, strategies designed to prevent basophil-mediated viral capture and transfer may be developed into a new form of therapy. IMPORTANCE Cell-associated HIV-1 infection has been proposed to play a pivotal role in the spread of HIV-1 infection. Here, we demonstrated that human blood-circulating granulocytes, particularly basophils, can capture HIV-1 and mediate viral trans-infection of CD4+ T cells. The expression of a variety of HIV-1 attachment factors, such as the C-type lectins, etc., facilitates viral capture and transfer. Intriguingly, the frequency of basophils in patients with different levels of CD4+ T counts remains fairly stable during the course of disease. Our results provide novel insights into the roles of granulocytes, particularly basophils, in HIV-1 dissemination. We suggest that strategies designed to prevent basophil-mediated viral capture and transfer may be a new direction for the development of anti-HIV therapy.
Collapse
|
17
|
Dynamic Micelles of Mannoside Glycolipids are more Efficient than Polymers for Inhibiting HIV-1 trans-Infection. Bioconjug Chem 2013; 24:1813-23. [DOI: 10.1021/bc4000806] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
18
|
Merbah M, Arakelyan A, Edmonds T, Ochsenbauer C, Kappes JC, Shattock RJ, Grivel JC, Margolis LB. HIV-1 expressing the envelopes of transmitted/founder or control/reference viruses have similar infection patterns of CD4 T-cells in human cervical tissue ex vivo. PLoS One 2012; 7:e50839. [PMID: 23236398 PMCID: PMC3516523 DOI: 10.1371/journal.pone.0050839] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 10/25/2012] [Indexed: 12/21/2022] Open
Abstract
Recently, it was found that 80% of sexual HIV-1 transmissions are established by a single virion/viral genome. To investigate whether the transmitted/founder (T/F) viruses have specific biological properties favoring sexual transmission, we inoculated human cervical tissue explants with isogenic HIV-1 viruses encoding Env sequences from T/F and control reference (C/R) HIV-1 variants as well as with full length T/F HIV-1 and compared their replication efficiencies, T cell depletion, and the activation status of infected cells. We found that all the HIV-1 variants were capable of transmitting infection to cervical tissue ex vivo and in this system preferentially replicate in activated CD4 T cells and deplete these cells. There was no difference in the biological properties of T/F and C/R HIV-1 variants as evaluated in ex vivo cervical tissue.
Collapse
Affiliation(s)
- Melanie Merbah
- Section of Intercellular Interactions, Program in Physical Biology, Eunice-Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Anush Arakelyan
- Section of Intercellular Interactions, Program in Physical Biology, Eunice-Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tara Edmonds
- University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | | - John C. Kappes
- University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | | | - Jean-Charles Grivel
- Section of Intercellular Interactions, Program in Physical Biology, Eunice-Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| | - Leonid B. Margolis
- Section of Intercellular Interactions, Program in Physical Biology, Eunice-Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
19
|
Dehuyser L, Schaeffer E, Chaloin O, Mueller CG, Baati R, Wagner A. Synthesis of Novel Mannoside Glycolipid Conjugates for Inhibition of HIV-1 Trans-Infection. Bioconjug Chem 2012; 23:1731-9. [DOI: 10.1021/bc200644d] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Laure Dehuyser
- Laboratory of Functional Chemo
Systems, CNRS-UdS UMR 7199, Faculté de Pharmacie, Université de Strasbourg, 74 route du Rhin,
67400 Illkirch, France
| | - Evelyne Schaeffer
- Laboratory of Immunology and
Therapeutic Chemistry, CNRS UPR 9021, Institut de Biologie Moléculaire et Cellulaire, 15 rue René
Descartes, 67000 Strasbourg, France
| | - Olivier Chaloin
- Laboratory of Immunology and
Therapeutic Chemistry, CNRS UPR 9021, Institut de Biologie Moléculaire et Cellulaire, 15 rue René
Descartes, 67000 Strasbourg, France
| | - Christopher G. Mueller
- Laboratory of Immunology and
Therapeutic Chemistry, CNRS UPR 9021, Institut de Biologie Moléculaire et Cellulaire, 15 rue René
Descartes, 67000 Strasbourg, France
| | - Rachid Baati
- Laboratory of Functional Chemo
Systems, CNRS-UdS UMR 7199, Faculté de Pharmacie, Université de Strasbourg, 74 route du Rhin,
67400 Illkirch, France
| | - Alain Wagner
- Laboratory of Functional Chemo
Systems, CNRS-UdS UMR 7199, Faculté de Pharmacie, Université de Strasbourg, 74 route du Rhin,
67400 Illkirch, France
| |
Collapse
|
20
|
Ahmed Z, Czubala M, Blanchet F, Piguet V. HIV impairment of immune responses in dendritic cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 762:201-38. [PMID: 22975877 DOI: 10.1007/978-1-4614-4433-6_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Dendritic cells and their subsets are diverse populations of immune cells in the skin and mucous membranes that possess the ability to sense the presence of microbes and orchestrate an efficient and adapted immune response. Dendritic cells (DC) have the unique ability to act as a bridge between the innate and adaptive immune responses. These cells are composed of a number of subsets behaving with preferential and specific features depending on their location and surrounding environment. Langerhans cells (LC) or dermal DC (dDC) are readily present in mucosal areas. Other DC subsets such as plasmacytoid DC (pDC), myeloid DC (myDC), or monocyte-derived DC (MDDC) are thought to be recruited or differentiated in sites of pathogenic challenge. Upon HIV infection, DC and their subsets are likely among the very first immune cells to encounter incoming pathogens and initiate innate and adaptive immune responses. However, as evidenced during HIV infection, some pathogens have evolved subtle strategies to hijack key cellular machineries essential to generate efficient antiviral responses and subvert immune responses for spread and survival.In this chapter, we review recent research aimed at investigating the involvement of DC subtypes in HIV transmission at mucosal sites, concentrating on HIV impact on cellular signalling and trafficking pathways in DC leading to DC-mediated immune response alterations and viral immune evasion. We also address some aspects of DC functions during the chronic immune pathogenesis and conclude with an overview of the current and novel therapeutic and prophylactic strategies aimed at improving DC-mediated immune responses, thus to potentially tackle the early events of mucosal HIV infection and spread.
Collapse
Affiliation(s)
- Zahra Ahmed
- Department of Dermatology and Wound Healing, Cardiff University School of Medicine, Cardiff, Wales, UK
| | | | | | | |
Collapse
|
21
|
Abstract
In the immune system, C-type lectins and CTLDs have been shown to act both as adhesion and as pathogen recognition receptors. The Dendritic cell-specific ICAM-3 grabbing non-integrin (DC-SIGN) and its homologs in human and mouse represent an important C-type lectin family. DC-SIGN contains a lectin domain that recognizes in a Ca2+-dependent manner carbohydrates such as mannose-containing structures present on glycoproteins such as ICAM-2 and ICAM-3. DC-SIGN is a prototype C-type lectin organized in microdomains, which have their role as pathogen recognition receptors in sensing microbes. Although the integrin LFA-1 is a counter-receptor for both ICAM-2 and ICAM-3 on DC, DC-SIGN is the high affinity adhesion receptor for ICAM-2/-3. While cell–cell contact is a primary function of selectins, collectins are specialized in recognition of pathogens. Interestingly, DC-SIGN is a cell adhesion receptor as well as a pathogen recognition receptor. As adhesion receptor, DC-SIGN mediates the contact between dendritic cells (DCs) and T lymphocytes, by binding to ICAM-3, and mediates rolling of DCs on endothelium, by interacting with ICAM-2. As pathogen receptor, DC-SIGN recognizes a variety of microorganisms, including viruses, bacteria, fungi and several parasites (Cambi et al. 2005). The natural ligands of DC-SIGN consist of mannose oligosaccharides or fucose-containing Lewis-type determinants. In this chapter, we shall focus on the structure and functions of DC-SIGN and related CTLDs in the recognition of pathogens, the molecular and structural determinants that regulate the interaction with pathogen-associated molecular patterns. The heterogeneity of carbohydrate residues exposed on cellular proteins and pathogens regulates specific binding of DC-expressed C-type lectins that contribute to the diversity of immune responses created by DCs (van Kooyk et al. 2003a; Cambi et al. 2005).
Collapse
|
22
|
Penicillium marneffei-stimulated dendritic cells enhance HIV-1 trans-infection and promote viral infection by activating primary CD4+ T cells. PLoS One 2011; 6:e27609. [PMID: 22110688 PMCID: PMC3217999 DOI: 10.1371/journal.pone.0027609] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2011] [Accepted: 10/20/2011] [Indexed: 11/23/2022] Open
Abstract
Penicillium marneffei (P. marneffei) is considered an indicator pathogen of AIDS, and the endemicity and clinical features of P. marneffei have been described. While, how the co-infection of P. marneffei exacerbate deterioration of the immune response remains poorly understood. Here we isolated P. marneffei from the cutaneous lesions of AIDS patients and analyzed its effects on HIV-1-dendritic cells (DCs) interaction. We demonstrated that the monocyte-derived dendritic cells (MDDCs) could be activated by both thermally dimorphic forms of P. marneffei for significantly promoting HIV-1 trans-infection of CD4+ T cells, while these activated MDDCs were refractory to HIV-1 infection. Mechanistically, P. marneffei-activated MDDCs endocytosed large amounts of HIV-1 and sequestrated the internalized viruses into tetrapasnin CD81+ compartments potentially for proteolysis escaping. The activated MDDCs increased expression of intercellular adhesion molecule 1 and facilitated the formation of DC-T-cell conjunctions, where much more viruses were recruited. Moreover, we found that P. marneffei-stimulated MDDCs efficiently activated resting CD4+ T cells and induced more susceptible targets for viral infection. Our findings demonstrate that DC function and its interaction with HIV-1 have been modulated by opportunistic pathogens such as P. marneffei for viral dissemination and infection amplification, highlighting the importance of understanding DC-HIV-1 interaction for viral immunopathogenesis elucidation.
Collapse
|