1
|
Trefulka M, Černocká H, Staroňová T, Ostatná V. Voltammetric analysis of glycoproteins containing sialylated and neutral glycans at pyrolytic graphite electrode. Bioelectrochemistry 2025; 163:108851. [PMID: 39637451 DOI: 10.1016/j.bioelechem.2024.108851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/12/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024]
Abstract
Recently, it was described that neutral glycans can be distinguished from those containing sialic acid at the mercury electrode after modification with osmium(VI) N,N,N',N'-tetramethylethylenediamine (Os(VI)tem). Our work shows the possibility of studying glycans and glycoproteins at pyrolytic graphite electrodes depending on thepresence of sialic acid. Short glycans, glycans released from glycoproteins, and glycoproteins themselves yielded similar voltammetric responses after their modification by Os(VI)tem. Os(VI)tem modified glycans and glycoproteins produced acouple of cathodic and anodic peaks. Changing peak heights and potentials of glycans and glycoproteins pointed out the presence of sialic acid. These findings could be utilized to improve glycoprotein sensing by chemical modification.
Collapse
Affiliation(s)
- Mojmír Trefulka
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics CAS, v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Hana Černocká
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics CAS, v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Tatiana Staroňová
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics CAS, v.v.i., Královopolská 135, 612 00 Brno, Czech Republic
| | - Veronika Ostatná
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics CAS, v.v.i., Královopolská 135, 612 00 Brno, Czech Republic.
| |
Collapse
|
2
|
Charnsatabut C, Suwanchaikasem P, Rattanapisit K, Iksen I, Pongrakhananon V, Bulaon CJI, Phoolcharoen W. Optimized expression of human interleukin-15 in Nicotiana benthamiana and in vitro assessment of its activity on human keratinocytes. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2025; 46:e00889. [PMID: 40235517 PMCID: PMC11997404 DOI: 10.1016/j.btre.2025.e00889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/16/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025]
Abstract
Human interleukin-15 (hIL-15) is a cytokine essential for immune modulation with therapeutic applications in cancer and chronic wound healing. Although hIL-15 is commercially available, large-scale production studies remain limited. With promising clinical trial results, demand for hIL-15 is expected to rise. Plant expression systems offer a sustainable, low-cost alternative for rapid biopharmaceutical production. In this study, we optimized hIL-15 expression in Nicotiana benthamiana and assessed its physicochemical properties and biological activity. We fused hIL-15 to the Fc domain of human IgG1 for efficient purification. Through optimization of the pre- and post-infiltration conditions, we achieved transient expression and recovery at 4 dpi, yielding 33.8 µg/g fresh weight. Peptide mapping confirmed 97 % overall sequence coverage of the primary structure. Treatment with plant-produced hIL-15-Fc effectively promoted human keratinocyte HaCaT cell proliferation and migration in vitro. These findings demonstrated the potential of plant-based platforms for producing therapeutic recombinant hIL-15 that support wound healing.
Collapse
Affiliation(s)
- Chalatorn Charnsatabut
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | - Iksen Iksen
- Department of Research and development, Provenedge Co. Ltd., Bangkok 10330, Thailand
| | - Varisa Pongrakhananon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Center of Excellence in Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Waranyoo Phoolcharoen
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
3
|
Shabo I, Nordling E, Abraham-Nordling M. Artificial intelligence prediction of carcinoembryonic antigen structure and interactions relevant for colorectal cancer. Biochem Biophys Rep 2025; 42:102024. [PMID: 40330074 PMCID: PMC12051046 DOI: 10.1016/j.bbrep.2025.102024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/08/2025] Open
Abstract
Carcinoembryonic antigen (CEA) is used as a biomarker for colorectal cancer. It is expressed during fetal development but in healthy adult cells the expression is low. Due to its size and the high degree of glycosylation, there are no structures available for mature CEA. By employing novel structure prediction methods, we aim to investigate CEA tertiary structure and interactions. Alphafold 3 server has increased the accuracy of structure predictions and allows for modelling of glycans in proteins and complexes. Models were created for a monomeric CEA, dimeric CEA and for CEA in complex with the antibody Tusamitamab. The structure of the monomeric glycosylated CEA exhibit two bends, one in the domain interface B1-A2 and one in the domain interface B2-A3. The dimer structure pairs in a parallel manner, with direct contacts in the N and the A2 domains of the two chains. The complex of CEA with Tusamitamab closely resembles the EM structure of the complex that was released after the training of Alphafold 3 was completed. Overall, the investigations give new angles to investigate for CEA. The predicted bend, primarily in the B2 and A3 domain interface, would allow for dimer formation of CEA from both the same cell as from adjacent cells and could help to explain the outstanding issue on how it can fulfil both tasks. The prediction of the antibody binding to CEA was accurate, the all-atom RMSD was 1.3 Å. This is encouraging for other antibody - protein complexes predictions as the complex structure was not part of the training set for Alphafold 3.
Collapse
Affiliation(s)
- Ivan Shabo
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Erik Nordling
- Swedish Orphan Biovitrum AB, Stockholm, 112 76, Sweden
| | | |
Collapse
|
4
|
Bassiouni W, Chan BYH, Roczkowsky A, Kranrod JW, Mahmud Z, Primeau JO, Cho WJ, Young HS, Seubert JM, Schulz R. Matrix metalloproteinase inhibition protects against junctophilin-2 proteolysis during doxorubicin-induced cardiotoxicity. Br J Pharmacol 2025. [PMID: 40375606 DOI: 10.1111/bph.70047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 02/25/2025] [Accepted: 03/11/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND AND PURPOSE Treatment of cancer patients with anthracyclines is known to cause dose-dependent cardiotoxicity through several mechanisms including enhanced oxidative stress, ultimately resulting in defective excitation-contraction coupling. Loss of junctophilin-2 (JPH-2), which tethers transverse tubules (T-tubules) to the sarcoplasmic reticulum, is a feature of doxorubicin-induced cardiotoxicity, yet the protease involved in unclear. As activation of matrix metalloproteinase-2 (MMP-2) is known to contribute to doxorubicin-induced cardiotoxicity, we investigated here the role of MMP-2 in JPH-2 proteolysis and defective calcium transients in it. EXPERIMENTAL APPROACH C57BL/6J mice were treated with doxorubicin for 4 weeks with or without the MMP inhibitor (doxycycline), MMP-2 preferring inhibitor (ONO-4817) or vehicle, and cardiac function was assessed using echocardiography. JPH-2 levels in ventricular extracts were measured. Calcium transients and JPH-2 levels were measured in neonatal rat ventricular cardiomyocytes treated with doxorubicin and ONO-4817. KEY RESULTS Both MMP inhibitors attenuated doxorubicin-induced cardiac systolic and diastolic dysfunction. Doxorubicin treatment resulted in JPH-2 cleavage in mouse hearts as evidenced by the appearance of lower molecular weight products of 63 and 25 kDa, which was prevented by MMP inhibitors. Loss of JPH-2 and impaired calcium transients were observed in neonatal rat ventricular cardiomyocytes treated with doxorubicin, while ONO-4817 attenuated these changes. In silico analysis predicted cleavage sites between JPH-2 MORN repeats and within its unstructured region. CONCLUSIONS AND IMPLICATIONS These results reveal that JPH-2 proteolysis is a consequence of MMP-2 activation and highlight the beneficial prophylactic action of two orally available MMP inhibitors in preventing doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Wesam Bassiouni
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Brandon Y H Chan
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Genomic Medicine, Alexion, AstraZeneca Rare Disease, Cambridge, Massachusetts, USA
| | - Andrej Roczkowsky
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joshua W Kranrod
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Zabed Mahmud
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joseph O Primeau
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Woo Jung Cho
- Cell and Tissue Imaging Center at St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Howard S Young
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - John M Seubert
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Richard Schulz
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
5
|
Kaufman B, Abu-Ahmad M, Radinsky O, Gharra E, Manko T, Bhattacharya B, Gologan D, Erlichman N, Meshel T, Nuta Y, Cooks T, Elkabets M, Ben-Baruch A, Porgador A. N-glycosylation of PD-L1 modulates the efficacy of immune checkpoint blockades targeting PD-L1 and PD-1. Mol Cancer 2025; 24:140. [PMID: 40346531 PMCID: PMC12065222 DOI: 10.1186/s12943-025-02330-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND The PD-L1/PD-1 pathway is crucial for immune regulation and has become a target in cancer immunotherapy. However, in order to improve patient selection for immune checkpoint blockade (ICB) therapies, better selection criteria are needed. This study explores how the N-glycosylation of PD-L1 affects its interaction with PD-1 and ICB efficacy, focusing on its four N-linked glycosylation sites: N35, N192, N200, and N219. METHODS Human PD-L1 glycosylation mutants-at each individual site or at all four sites together (Nx4)-were tested for their functional interaction with PD-1 using an artificial immune checkpoint reporter assay (IcAR-PD1). The blocking efficacy of anti-PD-L1 and anti-PD-1 antibodies was evaluated using human breast cancer cell lines (MDA-MB231 and MCF7), as well as A375 melanoma and A549 lung carcinoma cells expressing the glycosylation mutants. Results were validated through ex vivo activation and cytotoxicity assays using human CD8+ T cells. RESULTS The binding of the PD-L1N35A mutant to PD-1 was not effectively blocked by anti-PD-L1 and anti-PD-1 ICBs. In contrast, high blocking efficacy of PD-L1 binding to PD-1 was obtained at minimal ICB concentrations when PD-L1 did not express any glycosylation site (PD-L1Nx4 mutant). The PD-L1N35A mutant produced elevated levels of PD-L1 as a soluble (sPD-L1) and extracellular vesicles (EV)-bound molecule; in contrast, the PD-L1Nx4 mutant had lower sPD-L1 and EV levels. PD-L1 glycosylation status influenced the ability of PD-L1 interactions with PD-1 to down-regulate T-cell activation and cytotoxicity, with the PD-L1N35A mutant showing the lowest levels of T cell functions and the PD-L1Nx4 mutant the highest. CONCLUSIONS The N-glycosylation of PD-L1 at all four sites interferes with the ability of anti-PD-L1 and anti-PD-1 ICBs to block PD-L1 interactions with PD-1; in contrast, glycosylation at the N35 site enhances ICB blocking efficacy. These effects are connected to the ability of sPD-L1 to compete with ICB binding to PD-L1 or PD-1. Thus, assessing PD-L1 glycosylation, beyond expression levels, could improve patient stratification and outcomes.
Collapse
Affiliation(s)
- Bar Kaufman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Muhammad Abu-Ahmad
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Olga Radinsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Eman Gharra
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Tal Manko
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Baisali Bhattacharya
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Daniela Gologan
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Nofar Erlichman
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Tsipi Meshel
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Yoav Nuta
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Tomer Cooks
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel.
| | - Adit Ben-Baruch
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 6997801, Israel.
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, 84105, Israel.
| |
Collapse
|
6
|
Yan Z, Han J, Mi Z, Wang Z, Fu Y, Wang C, Dang N, Liu H, Zhang F. GPNMB disrupts SNARE complex assembly to maintain bacterial proliferation within macrophages. Cell Mol Immunol 2025; 22:512-526. [PMID: 40038549 PMCID: PMC12041529 DOI: 10.1038/s41423-025-01272-z] [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: 09/29/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
Xenophagy plays a crucial role in restraining the growth of intracellular bacteria in macrophages. However, the machinery governing autophagosome‒lysosome fusion during bacterial infection remains incompletely understood. Here, we utilize leprosy, an ideal model for exploring the interactions between host defense mechanisms and bacterial infection. We highlight the glycoprotein nonmetastatic melanoma protein B (GPNMB), which is highly expressed in macrophages from lepromatous leprosy (L-Lep) patients and interferes with xenophagy during bacterial infection. Upon infection, GPNMB interacts with autophagosomal-localized STX17, leading to a reduced N-glycosylation level at N296 of GPNMB. This modification promotes the degradation of SNAP29, thus preventing the assembly of the STX17-SNAP29-VAMP8 SNARE complex. Consequently, the fusion of autophagosomes with lysosomes is disrupted, resulting in inhibited cellular autophagic flux. In addition to Mycobacterium leprae, GPNMB deficiency impairs the proliferation of various intracellular bacteria in human macrophages, suggesting a universal role of GPNMB in intracellular bacterial infection. Furthermore, compared with their counterparts, Gpnmbfl/fl Lyz2-Cre mice presented decreased Mycobacterium marinum amplification. Overall, our study reveals a previously unrecognized role of GPNMB in host antibacterial defense and provides insights into its regulatory mechanism in SNARE complex assembly.
Collapse
Affiliation(s)
- Zhenzhen Yan
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Jinghong Han
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zihao Mi
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhenzhen Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yixuan Fu
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Chuan Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ningning Dang
- Department of Dermatology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Hong Liu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Furen Zhang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China.
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
7
|
Funayama R, Wang Y, Hosogane M, Kao W, Toyama S, Ohira M, Matsumoto M, Aizawa T, Kobayashi M, Karasawa H, Ohnuma S, Nakayama KI, Unno M, Nakayama K. Alternative Splicing of FBLN2 Generates a Prometastatic Extracellular Matrix in Gastrointestinal Cancers by Determining N-Glycosylation of Fibulin 2. Genes Cells 2025; 30:e70027. [PMID: 40400104 PMCID: PMC12095903 DOI: 10.1111/gtc.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/19/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025]
Abstract
Fibulin 2 (FBLN2) is an extracellular matrix glycoprotein. Exclusion of exon 9 of FBLN2 is one of the most recurrent splicing events across multiple types of cancer, but its functional relevance in cancer has remained unexplored. We here reveal that the exclusion of exon 9 of FBLN2 results in the loss of a single N-glycosylation site that leads to misfolding of the FBLN2 protein as well as to a reduction in both its stability and secretion efficiency. Indeed, the extracellular matrix of human colorectal cancer tissue exhibits a reduced abundance of FBLN2. This deficiency of FBLN2 together with a concomitant increase in the abundance of fibronectin 1 in the tumor microenvironment promotes the adhesion and migration of colorectal cancer cells. Our data thus suggest that the alternative splicing of FBLN2 exon 9 generates a prometastatic extracellular environment in cancer tissue by determining FBLN2 glycosylation.
Collapse
Affiliation(s)
- Ryo Funayama
- Department of Cell Proliferation, ART, Graduate School of MedicineTohoku UniversitySendaiJapan
- Anticancer Strategies Laboratory, Advanced Research InitiativeInstitute of Science TokyoTokyoJapan
| | - Yujue Wang
- Department of Cell Proliferation, ART, Graduate School of MedicineTohoku UniversitySendaiJapan
- Department of Cellular Function, Graduate School of Life SciencesTohoku UniversitySendaiJapan
| | - Masaki Hosogane
- Department of Cell Proliferation, ART, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Wei‐Chen Kao
- Department of Cell Proliferation, ART, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Shingo Toyama
- Department of Surgery, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Masahiro Ohira
- Department of Cell Proliferation, ART, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Masaki Matsumoto
- Department of Omics and Systems Biology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Takashi Aizawa
- Department of Surgery, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Minoru Kobayashi
- Department of Surgery, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Hideaki Karasawa
- Department of Surgery, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Shinobu Ohnuma
- Department of Surgery, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Keiichi I. Nakayama
- Anticancer Strategies Laboratory, Advanced Research InitiativeInstitute of Science TokyoTokyoJapan
- Department of Molecular and Cellular Biology, Medical Institute of BioregulationKyushu UniversityFukuokaJapan
| | - Michiaki Unno
- Department of Surgery, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Keiko Nakayama
- Department of Cell Proliferation, ART, Graduate School of MedicineTohoku UniversitySendaiJapan
- Research Infrastructure Management CenterInstitute of Science TokyoTokyoJapan
| |
Collapse
|
8
|
Ramírez-Salinas G, Shoshani L, Rosas-Trigueros JL, Huerta CS, Martínez-Archundia M. In silico studies provide new structural insights into trans-dimerization of β1 and β2 subunits of the Na+, K+-ATPase. PLoS One 2025; 20:e0321064. [PMID: 40299990 PMCID: PMC12040271 DOI: 10.1371/journal.pone.0321064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/02/2025] [Indexed: 05/01/2025] Open
Abstract
The Na+, K+-ATPase is an electrogenic transmembrane pump located in the plasma membrane of all animal cells. It is a dimeric protein composed of α and β subunits and has a third regulatory subunit (γ) belonging to the FXYD family. This pump plays a key role in maintaining low concentration of sodium and high concentration of potassium intracellularly. The α subunit is the catalytic one while the β subunit is important for the occlusion of the K+ ions and plays an essential role in trafficking of the functional αβ complex of Na+, K+-ATPase to the plasma membrane. Interestingly, the β1 and β2 (AMOG) isoforms of the β subunit, function as cell adhesion molecules in epithelial cells and astrocytes, respectively. Early experiments suggested a heterotypic adhesion for the β2. Recently, we reported a homotypic trans-interaction between β2-subunits expressed in CHO cells. In this work we use In Silico methods to analyze the physicochemical properties of the putative homophilic trans-dimer of β2 subunits and provide insights about the trans-dimerization interface stability. Our structural analysis predicts a molecular recognition mechanism of a trans-dimeric β2 - β2 subunit and permits designing experiments that will shed light upon possible homophilic interactions of β2 subunits in the nervous system.
Collapse
Affiliation(s)
- Gema Ramírez-Salinas
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| | - Liora Shoshani
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Jorge L. Rosas-Trigueros
- Laboratorio Transdisciplinario de Investigación enSistemas Evolutivos, ESCOM, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Christian Sosa Huerta
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - Marlet Martínez-Archundia
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
9
|
Suen TK, Al B, Ulas T, Reusch N, Bahrar H, Bekkering S, Bhat J, Kabelitz D, Schultze JL, van de Veerdonk FL, van Lennep JR, Riksen NP, Joosten LAB, Netea MG, Placek K. Human γδ T Cell Function Is Impaired Upon Mevalonate Pathway Inhibition. Immunology 2025. [PMID: 40264329 DOI: 10.1111/imm.13931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/17/2025] [Accepted: 04/06/2025] [Indexed: 04/24/2025] Open
Abstract
Vδ2 T cells, a predominant human peripheral γδ T cell population, are a promising candidate for the development of immunotherapies against cancer and infected cells. Aminobisphosphonate drugs, such as zoledronate, are commonly used to expand Vδ2 T cells. Yet, such in vitro generated cells have limited efficacy in the clinic. We found that despite inducing excessive proliferation of Vδ2 T cells, zoledronate impaired their effector function and caused the upregulation of the inhibitory receptor TIM3. This effect was due to the inhibition of mevalonate metabolism and dysregulation of downstream biological processes such as protein prenylation and intracellular signalling. In vitro and in vivo inhibition of mevalonate metabolism with zoledronate, statins, and 6-fluoromevalonate, as well as genetic deficiency of the mevalonate kinase, all resulted in compromised cytokine and cytotoxic molecule production by Vδ2 T cells. Impaired Vδ2 T cell function was accompanied by transcriptome and kinome changes. Our findings reveal the importance of mevalonate metabolism for the proper functioning of Vδ2 T cells. This observation provides important considerations for improving their therapeutic use and has repercussions for patients with statin or aminobisphosphonate treatments.
Collapse
Affiliation(s)
- Tsz Kin Suen
- Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Burcu Al
- Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Thomas Ulas
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Bonn, Germany
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Nico Reusch
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Bonn, Germany
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jaydeep Bhat
- Institute of Immunology, University of Kiel, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, University of Kiel, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Joachim L Schultze
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics at the DZNE and the University of Bonn, Bonn, Germany
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Frank L van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Katarzyna Placek
- Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
10
|
Barroso da Silva FL, Paco K, Laaksonen A, Ray A. Biophysics of SARS-CoV-2 spike protein's receptor-binding domain interaction with ACE2 and neutralizing antibodies: from computation to functional insights. Biophys Rev 2025; 17:309-333. [PMID: 40376405 PMCID: PMC12075047 DOI: 10.1007/s12551-025-01276-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 01/24/2025] [Indexed: 05/18/2025] Open
Abstract
The spike protein encoded by the SARS-CoV-2 has become one of the most studied macromolecules in recent years due to its central role in COVID-19 pathogenesis. The spike protein's receptor-binding domain (RBD) directly interacts with the host-encoded receptor protein, ACE2. This review critically examines computational insights into RBD's interaction with ACE2 and with therapeutic antibodies designed to interfere with this interaction. We begin by summarizing insights from early computational studies on pre-pandemic SARS-CoV-1 RBD interactions and how these early studies shaped the understanding of SARS-CoV-2. Next, we highlight key theoretical contributions that revealed the molecular mechanisms behind the binding affinity of SARS-CoV-2 RBD against ACE2, and the structural changes that have enhanced the infectivity of emerging variants. Special attention is given to the "RBD charge rule", a predictive framework for determining variant infectivity based on the electrostatic properties of the RBD. Towards applying the computational insights to therapy, we discuss a multiscale computational protocol for optimizing monoclonal antibodies to improve binding affinity across multiple spike protein variants, including representatives from the Omicron family. Finally, we explore how these insights can inform the development of future vaccines and therapeutic interventions for combating future coronavirus diseases.
Collapse
Affiliation(s)
- Fernando Luís Barroso da Silva
- Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av Prof Zeferino Vaz, S/no, Ribeirão Preto, São Paulo BR-14040-903 Brazil
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Engineering Building I (EB1), Raleigh, NC 27695-7905 USA
| | - Karen Paco
- Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr., Claremont, CA 91711 USA
| | - Aatto Laaksonen
- Department of Chemistry, Arrhenius Laboratory, Stockholm University, Svante Arrhenius Väg 8, 106 91 Stockholm, Sweden
- State Key Laboratory of Materials-Oriented and Chemical Engineering, Nanjing Tech University, NO.30 Puzhu Road(S), Nanjing, 210009 People’s Republic of China
- Department of Engineering Sciences and Mathematics, Division of Energy Science, Luleå University of Technology, Laboratorievägen 14, 97187 Luleå, Sweden
- Centre of Advanced Research in Bionanoconjugates and Biopolymers, Petru Poni Institute of Macromolecular Chemistry, Aleea Grigore Ghica-Voda, 41A, 700487 Iasi, Romania
| | - Animesh Ray
- Riggs School of Applied Life Sciences, Keck Graduate Institute, 535 Watson Dr., Claremont, CA 91711 USA
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125 USA
| |
Collapse
|
11
|
Kobayashi D, Hiono T, Arakawa H, Kaji H, Ohkawara A, Ichikawa T, Ban H, Isoda N, Sakoda Y. Deglycosylation and truncation in the neuraminidase stalk are functionally equivalent in enhancing the pathogenicity of a high pathogenicity avian influenza virus in chickens. J Virol 2025; 99:e0147824. [PMID: 39950775 PMCID: PMC11915841 DOI: 10.1128/jvi.01478-24] [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: 08/23/2024] [Accepted: 12/21/2024] [Indexed: 03/19/2025] Open
Abstract
Influenza A viruses with fewer amino acids in the neuraminidase (NA) stalk domain are primarily isolated from chickens rather than wild ducks, indicating that a shortened NA stalk is considered an adaptation marker of avian influenza viruses (AIVs) to chickens. Experimental passages of an H7N7 nonpathogenic AIV (rgVac2-P0) in chickens resulted in a highly pathogenic variant (Vac2-P3L4) with a 34-amino-acid deletion in the NA stalk, encompassing five potential N-glycosylation sites. To investigate how amino acid truncation and deglycosylation in the NA stalk contribute to increased pathogenicity, a virus with glycosylation-deficient mutations at these sites (rgVac2-P3L4/P0NAΔGlyco) was constructed. Contrary to expectations, chickens inoculated with rgVac2-P3L4/P0NAΔGlyco exhibited variable clinical outcomes, attributed to the genetic instability of the virus. A single mutation stabilized the virus, and the mutant (rgVac2-P3L4/P0NAΔGlyco-Y65H) resulted in higher pathogenicity compared with a virus with restored glycosylation (rgVac2-P3L4/P0NA-Y65H). Glycan occupancy analysis revealed 3-4 glycans at the five potential sites. In functional analysis, glycosylation-deficient mutants, similar to the short-stalk NA virus, showed significantly reduced erythrocyte elution activity. Additionally, mutational analysis indicated variable contributions of N-glycans to elution activity across the sites. Moreover, the functionally most contributing sites of the five potential N-glycosylation motifs were consistently included in the amino acid deletions of the stalk-truncated NA in N7-subtyped field isolates, despite the varying truncation position or length. These findings suggest that the loss of glycosylation is functionally equivalent to a reduction in amino acids, and it plays a crucial role in enhancing pathogenicity in chickens and affecting NA function.IMPORTANCEAvian influenza poses significant economic challenges to the poultry industry and presents potential risks to human health. Understanding the molecular mechanisms that facilitate the emergence of chicken-adapted avian influenza viruses (AIVs) from non-pathogenic duck-origin influenza viruses is crucial for improving AIV monitoring systems in poultry and controlling this disease. Amino acid deletions in the neuraminidase (NA) stalk domain serve as one of the molecular markers for AIV adaptation to Galliformes. This study highlights the critical role of N-glycosylation in the NA stalk domain in the pathogenesis of high pathogenicity avian influenza viruses in chickens. The findings propose a novel theory that the loss of glycosylation at the NA stalk domain, rather than a reduction in stalk length, is responsible for both NA function and increased virus pathogenicity in chickens.
Collapse
Affiliation(s)
- Daiki Kobayashi
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takahiro Hiono
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Hokkaido, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hiromu Arakawa
- Systems Biology Division, Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Hiroyuki Kaji
- Systems Biology Division, Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, Japan
| | - Ayako Ohkawara
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takaya Ichikawa
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Microbiology and Immunology, Faculty of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hinako Ban
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Norikazu Isoda
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Hokkaido, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yoshihiro Sakoda
- Laboratory of Microbiology, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Hokkaido, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
12
|
Yanova M, Stepanova E, Maltseva D, Tonevitsky A. CD44 variant exons induce chemoresistance by modulating cell death pathways. Front Cell Dev Biol 2025; 13:1508577. [PMID: 40114966 PMCID: PMC11924683 DOI: 10.3389/fcell.2025.1508577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/03/2025] [Indexed: 03/22/2025] Open
Abstract
Cancer chemoresistance presents a challenge in oncology, often leading to treatment failure and disease progression. CD44, a multifunctional cell surface glycoprotein, has garnered attention for its involvement in various aspects of cancer biology. Through alternative splicing, CD44 can form isoforms with the inclusion of only standard exons, typical for normal tissue, or with the addition of variant exons, frequently expressed in cancer tissue and associated with chemoresistance. The functions of CD44 involved in regulation of cancer signaling pathways are being actively studied, and the significance of specific variant exons in modulating cell death pathways, central to the response of cancer cells to chemotherapy, begins to become apparent. This review provides a comprehensive analysis of the association of CD44 variant exons/total CD44 with clinical outcomes of patients undergoing chemotherapy. The role of CD44 variant exons v6, v9 and others with a significant effect on patient chemotherapy outcomes by means of key cellular death pathways such as apoptosis, ferroptosis and autophagy modulation is further identified, and their impact on drug resistance is highlighted. An overview of clinical trials aimed at targeting variant exon-containing isoforms is provided, and possible directions for further development of CD44-targeted therapeutic strategies are discussed.
Collapse
Affiliation(s)
- Maria Yanova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Evgeniya Stepanova
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Diana Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
13
|
Hadi AF, Arta RK, Kushima I, Egawa J, Watanabe Y, Ozaki N, Someya T. Association Analysis of Rare CNTN5 Variants With Autism Spectrum Disorder in a Japanese Population. Neuropsychopharmacol Rep 2025; 45:e12527. [PMID: 39887962 PMCID: PMC11781355 DOI: 10.1002/npr2.12527] [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: 09/19/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Contactin-5 (CNTN5), a neural adhesion molecule involved in synaptogenesis and synaptic maturation in the auditory pathway, has been associated with the pathophysiology of autism spectrum disorder (ASD), particularly hyperacusis. To investigate the role of rare CNTN5 variants in ASD susceptibility, we performed resequencing and association analysis in a Japanese population. METHODS We resequenced the CNTN5 coding regions in 302 patients with ASD and prioritized rare putatively damaging variants. The prioritized variants were then genotyped in 313 patients with ASD and 1065 controls. Subsequently, we conducted an association study of selected variants with ASD in 614 patients with ASD and 61 057 controls. Clinical data were reviewed for patients carrying prioritized variants. RESULTS Through resequencing, we prioritized three rare putatively damaging missense variants (W69G, I227L, and L1000S) in patients with ASD. Although we found a nominally significant association between the I227L variant and ASD, it did not remain significant after post hoc correction. Hyperacusis was found in three out of nine patients carrying prioritized variants. CONCLUSION This study does not provide evidence for the contribution of rare CNTN5 variants to the genetic etiology of ASD in the Japanese population.
Collapse
Affiliation(s)
- Abdul Fuad Hadi
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Reza K. Arta
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Itaru Kushima
- Department of PsychiatryNagoya University Graduate School of MedicineNagoyaAichiJapan
- Medical Genomics CenterNagoya University HospitalNagoyaAichiJapan
| | - Jun Egawa
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Yuichiro Watanabe
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
- Department of PsychiatryUonuma Kikan HospitalNiigataJapan
| | - Norio Ozaki
- Pathophysiology of Mental DisordersNagoya University Graduate School of MedicineNagoyaAichiJapan
| | - Toshiyuki Someya
- Department of Psychiatry, School of Medicine, and Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| |
Collapse
|
14
|
Dasari S, Kalyaanamoorthy S. Impact of Phosphorylation and O-GlcNAcylation on the Binding Affinity of R4 Tau Peptide to Microtubule and Its Conformational Preference upon Dissociation. J Chem Inf Model 2025; 65:1570-1584. [PMID: 39871444 DOI: 10.1021/acs.jcim.4c02109] [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: 01/29/2025]
Abstract
Tau is a microtubule (MT)-associated protein that binds to and stabilizes the MTs of neurons. Due to its intrinsically disordered nature, it undergoes several post-translational modifications (PTMs) that are intricately linked to both the physiological and pathophysiological roles of Tau. Prior research has shown phosphorylation and O-GlcNAcylation to have contrasting effects on Tau aggregation; however, the precise molecular mechanisms and potential synergistic effects of these modifications remain elusive. In this article, we study the impact of phosphorylation at S352, and S356, as well as the phosphorylation of O-GlcNAcylation at S356, individually and in combination, on the binding of the R4 (336-367) peptide with MTs by performing classical molecular dynamics (MD) simulations. By analyzing the binding free energies of the Tau-MT complex, we found that both individual and combined phosphorylation at S352 and S356 sites decreased the affinity of the R4 peptide toward MT. Surprisingly, O-GlcNAcylation, a likely neuroprotective modification, at S356 also decreased the binding affinity of Tau to MT similar to the single phosphorylation systems (pS352 or pS356) but was observed to maintain major interactions with MT comparable to unmodified R4. Additionally, we investigated the impact of phosphorylation at both sites and the interplay between phosphorylation at S352 and O-GlcNAcylation at S356, which showed that the latter preserved the interactions and affinity of the Tau with MT better than dual phosphorylation, though still not as effectively as single phosphorylation. These findings suggest that O-GlcNAcylation at residue S356 has a moderate destabilizing effect. We also performed replica-exchange MD simulations of the R4 peptide to understand the changes in conformational preferences upon phosphorylation, O-GlcNAcylation, and a combination of both modifications. Both individual and combined phosphorylation of R4 peptide at S352, and S356, sites induced salt-bridge interactions with positively charged side chains of lysine and arginine amino acids. However, O-GlcNAcylation at S356 induced secondary structural changes on the R4 peptide, leading to the formation of a β-sheet structure, consistent with previous experimental observations. Interestingly, simultaneous phosphorylation at S352 and the phosphorylation of O-GlcNAcylation at S356 resulted in conformations promoting salt-bridges and β-sheets. Thus, our study provides atomistic insights into the impact of PTMs on the binding of Tau peptide to MT and its conformational preferences upon dissociation.
Collapse
Affiliation(s)
- Sathish Dasari
- Department of Chemistry, Faculty of Science, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Subha Kalyaanamoorthy
- Department of Chemistry, Faculty of Science, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
- Waterloo Artificial Intelligence Institute, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
- Waterloo Institute of Nanotechnology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
15
|
Geng L, Yi X, Lin Y, Abulimiti X, Jin L, Yu J, Xu A. Site-specific analysis and functional characterization of N-linked glycosylation for β-Klotho protein. Int J Biol Macromol 2025; 289:138846. [PMID: 39701265 DOI: 10.1016/j.ijbiomac.2024.138846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 12/21/2024]
Abstract
β-Klotho (KLB), a type I transmembrane protein, serves as an obligate co-receptor determining the tissue-specific actions of endocrine fibroblast growth factors (FGFs). Despite accumulative evidence suggesting the occurrence of N-glycosylation in the KLB protein, the precise N-glycosites, glycoforms, and the impacts of N-glycosylation on the expression and function of the KLB protein remain unexplored. Employing a mass spectrometry-based approach, a total of 12 N-glycosites displaying heterogeneous site occupancy and glycoforms were identified within the extracellular region of the recombinant human KLB protein. Molecular simulation revealed negligible impact of these N-glycans on the overall structure of the KLB protein. However, both pharmacological inhibition of N-glycosylation and mutagenesis targeting N-glycosites reduced mature KLB protein content without impacting KLB mRNA synthesis in cells, underscoring the critical role of N-glycosylation in maintaining the stability of the KLB protein. Further studies revealed that the underglycosylated KLB mutant underwent rapid degradation via both lysosomal and proteasomal pathways and was unable to be efficiently trafficked to the plasma membrane, leading to impaired FGF21 signaling transduction. Collectively, multisite N-glycosylation is essential for the stability and cell surface localization of the KLB protein, representing a novel modulatory mechanism of endocrine FGF signaling.
Collapse
Affiliation(s)
- Leiluo Geng
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China.
| | - Xinyao Yi
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China
| | - Ying Lin
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China
| | - Xiayidan Abulimiti
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China
| | - Leigang Jin
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China
| | - Jiasui Yu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China; Department of Medicine, School of Clinical Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, 21 Sassoon Road, Pokfulam 999077, Hong Kong, China.
| |
Collapse
|
16
|
Li XL, Yan ZS, Ma YQ, Ding HM. Impact of Glycosylation of Apolipoprotein D on Its Interaction with Gold Nanoparticles: Insights from Molecular Dynamics Simulations. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4490-4501. [PMID: 39772418 DOI: 10.1021/acsami.4c16685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Efficient delivery of nanoparticles (NPs) as carriers for biochemical substances is crucial in various biomedical applications. In this study, we systematically investigate the interactions between glycosylated and nonglycosylated forms of Apolipoprotein D (ApoD) with gold nanoparticles (AuNPs) functionalized with different polymer coatings, including polyethylene glycol (PEG) and zwitterionic polymers. Using all-atom molecular dynamics simulations, we demonstrate that glycosylation significantly enhances the adsorption behavior of ApoD on AuNP surfaces, with the extent of this enhancement being dependent on the type (especially the charge property) of the polymer coatings. Notably, while zwitterionic polymers exhibit strong resistance to protein adsorption in their nonglycosylated form, this antifouling capability is diminished when glycosylation is present. Further, our findings reveal that glycosylation not only strengthens the binding energy of proteins but also alters the hydration dynamics at the NP-protein interface. Overall, this study provides a deeper understanding of the role of glycosylation in modulating protein-nanoparticle interactions, which is essential for the design of more effective nanomaterials for precision medicine.
Collapse
Affiliation(s)
- Xiao-Lei Li
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Zeng-Shuai Yan
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yu-Qiang Ma
- National Laboratory of Solid State Microstructures and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
- Jiangsu Physical Science Research Center, Nanjing 210093, China
| | - Hong-Ming Ding
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| |
Collapse
|
17
|
Jayaprakash NG, Sarkar DK, Surolia A. Atomic visualization of flipped-back conformations of high mannose glycans interacting with cargo lectins: An MD simulation perspective. Proteins 2025; 93:112-133. [PMID: 37465933 DOI: 10.1002/prot.26556] [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: 05/31/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023]
Abstract
Protein-carbohydrate interactions play a crucial role in mediating several biomolecular recognition events. We attempt to unravel its intricacies by understanding how carbohydrate-binding proteins interpret the glycan code. We aim to decipher lectin-mediated recognition in the endoplasmic reticulum (ER), which plays a crucial role in ER-mediated quality control (ER-QC). The ER-QC functions in three phases-protein folding, transport, and degradation. Altered protein QC leads to ER-related storage disorders. Cargo transport proteins-Ergic53 and Vip36-necessary for maintaining cellular homeostasis-are our primary focus. They recognize monoglucosylated/high mannose N-glycans on the folded glycoproteins. This article reports on the first dynamic investigation of the ER cargo lectins in complex with the high mannose glycans using an advanced sampling technique-replica exchange molecular dynamics to decipher the inherent conformational heterogeneity and the binding mechanism. The study involves simulations for the proteins complexed with three high mannose glycans-Man8B, Man9, and mono-glucosylated glycan. The recognition process is captured using MD simulations to achieve mechanistic insights and characterize the dynamics of glycans in their native and bound states via dihedral angle analysis. Results indicate that the flipped conformation of the glycans was crucial in differentiating their interaction with the proteins. Similar conformers of the glycans are preferred for Ergic53 and Vip36 in their glycan recognition events. Ergic53 preferred Man8B while it was Man9 for Vip36, in coherence with the previous experimental reports. These simulations provide a computational microscopic purview of the mechanism at both spatial and temporal scales. The results correlate with the published experimental data on the specificities of these lectins.
Collapse
Affiliation(s)
| | | | - Avadhesha Surolia
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| |
Collapse
|
18
|
Christofi E, O’Hanlon M, Curtis R, Barman A, Keen J, Nagy T, Barran P. Hybrid Mass Spectrometry Applied across the Production of Antibody Biotherapeutics. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2025; 36:44-57. [PMID: 39573914 PMCID: PMC11697328 DOI: 10.1021/jasms.4c00253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/12/2024] [Accepted: 10/01/2024] [Indexed: 01/02/2025]
Abstract
Post expression from the host cells, biotherapeutics undergo downstream processing steps before final formulation. Mass spectrometry and biophysical characterization methods are valuable for examining conformational and stoichiometric changes at these stages, although typically not used in biomanufacturing, where stability is assessed via bulk property studies. Here we apply hybrid MS methods to understand how solution condition changes impact the structural integrity of a biopharmaceutical across the processing pipeline. As an exemplar product, we use the model IgG1 antibody, mAb4. Flexibility, stability, aggregation propensity, and bulk properties are evaluated in relation to perfusion media, purification stages, and formulation solutions. Comparisons with Herceptin, an extensively studied IgG1 antibody, were conducted in a mass spectrometry-compatible solution. Despite presenting similar charge state distributions (CSD) in native MS, mAb4, and Herceptin show distinct unfolding patterns in activated ion mobility mass spectrometry (aIM-MS) and differential scanning fluorimetry (DSF). Herceptin's greater structural stability and aggregation onset temperature (Tagg) are attributed to heavier glycosylation and kappa-class light chains, unlike the lambda-class light chains in mAb4. Hydrogen-deuterium exchange mass spectrometry (HDX-MS) revealed that mAb4 undergoes substantial structural changes during purification, marked by high flexibility, low melting temperature (Tm), and prevalent repulsive protein-protein interactions but transitions to a compact and stable structure in high-salt and formulated environments. Notably, in formulation, the third constant domain (CH3) of the heavy chain retains flexibility and is a region of interest for aggregation. Future work could translate features of interest from comprehensive studies like this to targeted approaches that could be utilized early in the development stage to aid in decision-making regarding targeted mutations or to guide the design space of bioprocesses and formulation choices.
Collapse
Affiliation(s)
- Emilia Christofi
- Michael
Barber Centre for Collaborative Mass Spectrometry, MBCCMS, Princess Street, Manchester M17DN, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, Princess Street, Manchester M17DN, U.K.
| | - Mark O’Hanlon
- Manchester
Institute of Biotechnology, University of
Manchester, Princess Street, Manchester M17DN, U.K.
| | - Robin Curtis
- Manchester
Institute of Biotechnology, University of
Manchester, Princess Street, Manchester M17DN, U.K.
| | - Arghya Barman
- FUJIFILM
Diosynth Biotechnologies, Belasis Ave, Stockton-on-Tees, Billingham TS23 1LH, U.K.
| | - Jeff Keen
- FUJIFILM
Diosynth Biotechnologies, Belasis Ave, Stockton-on-Tees, Billingham TS23 1LH, U.K.
| | - Tibor Nagy
- FUJIFILM
Diosynth Biotechnologies, Belasis Ave, Stockton-on-Tees, Billingham TS23 1LH, U.K.
| | - Perdita Barran
- Michael
Barber Centre for Collaborative Mass Spectrometry, MBCCMS, Princess Street, Manchester M17DN, U.K.
- Manchester
Institute of Biotechnology, University of
Manchester, Princess Street, Manchester M17DN, U.K.
| |
Collapse
|
19
|
Silva MLS. Lectin-modified drug delivery systems - Recent applications in the oncology field. Int J Pharm 2024; 665:124685. [PMID: 39260750 DOI: 10.1016/j.ijpharm.2024.124685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Chemotherapy with cytotoxic drugs remains the core treatment for cancer but, due to the difficulty to find general and usable biochemical differences between cancer cells and normal cells, many of these drugs are associated with lack of specificity, resulting in side effects and collateral cytotoxicity that impair patients' adherence to therapy. Novel cancer treatments in which the cytotoxic effect is maximized while adverse effects are reduced can be implemented by developing targeted therapies that exploit the specific features of cancer cells, such as the typical expression of aberrant glycans. Modification of drug delivery systems with lectins is one of the strategies to implement targeted chemotherapies, as lectins are able to specifically recognize and bind to cancer-associated glycans expressed at the surface of cancer cells, guiding the drug treatment towards these cells and not affecting healthy ones. In this paper, recent advances on the development of lectin-modified drug delivery systems for targeted cancer treatments are thoroughly reviewed, with a focus on their properties and performance in diverse applications, as well as their main advantages and limitations. The synthesis and analytical characterization of the cited lectin-modified drug delivery systems is also briefly described. A comparison with free-drug treatments and with antibody-modified drug delivery systems is presented, emphasizing the advantages of lectin-modified drug delivery systems. Main constraints and potential challenges of lectin-modified drug delivery systems, including key difficulties for clinical translation of these systems, and the required developments in this area, are also signalled.
Collapse
Affiliation(s)
- Maria Luísa S Silva
- Centro de Estudos Globais, Universidade Aberta, Rua da Escola Politécnica 147, 1269-001 Lisboa, Portugal.
| |
Collapse
|
20
|
Dey R, Taraphder S. Molecular Modeling of Glycosylated Catalytic Domain of Human Carbonic Anhydrase IX. J Phys Chem B 2024; 128:11054-11068. [PMID: 39487784 DOI: 10.1021/acs.jpcb.4c03514] [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: 11/04/2024]
Abstract
Glycans exhibit significant structural diversity due to the flexibility of glycosidic bonds linking their constituent monosaccharides and the formation of numerous hydrogen bonds. The present work searches a simulated ensemble of glycan chain conformations attached to the catalytic domain of N-glycosylated human carbonic anhydrase IX (HCA IX-c) to identify conformations pointed away or back-folded toward the protein surface guided by different amino acid residues. A series of classical molecular dynamics (MD) simulation studies for a total of 30 μs followed by accelerated MD simulations for a total of 2 μs have been performed using two different force fields to capture varying degrees of fluctuations of both glycan chain and HCA IX. From the underlying free energy profile and kinetics derived using hidden Markov state model, several stable glycan orientations are identified that extend away from the protein surface and convert among each other with rate constants of the order 107-1010 S-1. Most importantly, we have identified a rare glycan conformation which reaches close to a catalytically important amino acid residue, Glu-106. We further enlist the protein residues that couple such less frequent event of the glycan chain back-folding toward the surface of the protein.
Collapse
Affiliation(s)
- Ritwika Dey
- Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, India
| | - Srabani Taraphder
- Department of Chemistry, Indian Institute of Technology, Kharagpur 721302, India
| |
Collapse
|
21
|
Kumar A, Duffieux F, Gagnaire M, Rapisarda C, Bertrand T, Rak A. Structural insights into epitope-paratope interactions of a monoclonal antibody targeting CEACAM5-expressing tumors. Nat Commun 2024; 15:9377. [PMID: 39477960 PMCID: PMC11525548 DOI: 10.1038/s41467-024-53746-9] [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: 08/04/2023] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) are overexpressed in some tumor types. The antibody-drug conjugate tusamitamab ravtansine specifically recognizes the A3-B3 domains of human CEACAM5 (hCEACAM5). To understand this specificity, here we map the epitope-paratope interface between the A3-B3 domains of hCEACAM5 (hCEACAM5A3-B3) and the antigen-binding fragment of tusamitamab (tusa Fab). We use hydrogen/deuterium exchange mass spectrometry to identify the tusa Fab paratope, which involves heavy chain (HC) residues 101-109 and light chain residues 48-54 and 88-104. Using surface plasmon resonance, we demonstrate that alanine variants of HC residues 96-108 abolish binding to hCEACAM5, suggesting that these residues are critical for tusa-Fab-antigen complex formation. The cryogenic electron microscopy structure of the hCEACAM5A3-B3- tusa Fab complex (3.11 Å overall resolution) reveals a discontinuous epitope involving residues in the A3-B3 domains and an N-linked mannose at residue Asn612. Conformational constraints on the epitope-paratope interface enable tusamitamab to target hCEACAM5A3-B3 and distinguish CEACAM5 from other CEACAMs.
Collapse
Affiliation(s)
- Anand Kumar
- Integrated Drug Discovery, Sanofi R&D, Paris, France
| | | | | | | | | | - Alexey Rak
- Integrated Drug Discovery, Sanofi R&D, Paris, France.
| |
Collapse
|
22
|
Paul R, Chakrabarty A, Samanta S, Dey S, Pandey R, Maji S, Pezacki AT, Chang CJ, Datta R, Gupta A. Leishmania major-induced alteration of host cellular and systemic copper homeostasis drives the fate of infection. Commun Biol 2024; 7:1226. [PMID: 39349621 PMCID: PMC11442737 DOI: 10.1038/s42003-024-06716-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 08/12/2024] [Indexed: 10/04/2024] Open
Abstract
Copper plays a key role in host-pathogen interaction. We find that during Leishmania major infection, the parasite-harboring macrophage regulates its copper homeostasis pathway in a way to facilitate copper-mediated neutralization of the pathogen. Copper-ATPase ATP7A transports copper to amastigote-harboring phagolysosomes to induce stress on parasites. Leishmania in order to evade the copper stress, utilizes a variety of manipulative measures to lower the host-induced copper stress. It induces deglycosylation and degradation of host-ATP7A and downregulation of copper importer, CTR1 by cysteine oxidation. Additionally, Leishmania induces CTR1 endocytosis that arrests copper uptake. In mouse model of infection, we report an increase in systemic bioavailable copper in infected animals. Heart acts as the major organ for diverting its copper reserves to systemic circulation to fight-off infection by downregulating its CTR1. Our study explores reciprocal mechanism of manipulation of host copper homeostasis pathway by macrophage and Leishmania to gain respective advantages in host-pathogen interaction.
Collapse
Affiliation(s)
- Rupam Paul
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India.
| | - Adrija Chakrabarty
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Suman Samanta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Swastika Dey
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Raviranjan Pandey
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Saptarshi Maji
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Aidan T Pezacki
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Christopher J Chang
- Department of Chemistry, University of California Berkeley, Berkeley, CA, USA
| | - Rupak Datta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Arnab Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India.
| |
Collapse
|
23
|
Ding W, Chen X, Sun Z, Luo J, Wang S, Lu Q, Ma J, Zhao C, Chen FE, Xu C. A Radical Activation Strategy for Versatile and Stereoselective N-Glycosylation. Angew Chem Int Ed Engl 2024; 63:e202409004. [PMID: 38837495 DOI: 10.1002/anie.202409004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/07/2024]
Abstract
Previous N-glycosylation approaches have predominately involved acidic conditions, facing challenges of low stereoselectivity and limited scope. Herein, we introduce a radical activation strategy that enables versatile and stereoselective N-glycosylation using readily accessible glycosyl sulfinate donors under basic conditions and exhibits exceptional tolerance towards various N-aglycones containing alkyl, aryl, heteroaryl and nucleobase functionalities. Preliminary mechanistic studies indicate a pivotal role of iodide, which orchestrates the formation of a glycosyl radical from the glycosyl sulfinate and subsequent generation of the key intermediate, a configurationally well-defined glycosyl iodide, which is subsequently attacked by an N-aglycone in a stereospecific SN2 manner to give the desired N-glycosides. An alternative route involving the coupling of a glycosyl radical and a nitrogen-centered radical is also proposed, affording the exclusive 1,2-trans product. This novel approach promises to broaden the synthetic landscape of N-glycosides, offering a powerful tool for the construction of complex glycosidic structures under mild conditions.
Collapse
Affiliation(s)
- Wenyan Ding
- Institute of Pharmaceutical Science and Technology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
- Qingyuan Innovation Laboratory, Quanzhou, 362801, China
| | - Xinyu Chen
- Institute of Pharmaceutical Science and Technology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zuyao Sun
- Institute of Pharmaceutical Science and Technology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jiaxin Luo
- Institute of Pharmaceutical Science and Technology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Shiping Wang
- National Engineering Research Center of Chemical Fertilizer Catalyst, College of Chemical Engineering, Fuzhou University, Fuzhou, 350108, China
| | - Qingqing Lu
- Institute of Pharmaceutical Science and Technology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jialu Ma
- Institute of Pharmaceutical Science and Technology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Chongxin Zhao
- Jiangsu Jiyi New Material CO., LTD, Xuzhou, 221700, China
| | - Fen-Er Chen
- Institute of Pharmaceutical Science and Technology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai, 200433, China
- Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Fudan University, Shanghai, 200433, China
| | - Chunfa Xu
- Institute of Pharmaceutical Science and Technology, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| |
Collapse
|
24
|
Goecker ZC, Burke MC, Remoroza CA, Liu Y, Mirokhin YA, Sheetlin SL, Tchekhovskoi DV, Yang X, Stein SE. Variation of Site-Specific Glycosylation Profiles of Recombinant Influenza Glycoproteins. Mol Cell Proteomics 2024; 23:100827. [PMID: 39128790 PMCID: PMC11417209 DOI: 10.1016/j.mcpro.2024.100827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 07/08/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024] Open
Abstract
This work presents a detailed determination of site-specific N-glycan distributions of the recombinant influenza glycoproteins hemagglutinin (HA) and neuraminidase. Variation in glycosylation among recombinant glycoproteins is not predictable and can depend on details of the biomanufacturing process as well as details of protein structure. In this study, recombinant influenza proteins were analyzed from eight strains of four different suppliers. These include five HA and three neuraminidase proteins, each produced from a HEK293 cell line. Digestion was conducted using a series of complex multienzymatic methods designed to isolate glycopeptides containing single N-glycosylated sites. Site-specific glycosylation profiles of intact glycopeptides were produced using a recently developed method and comparisons were made using spectral similarity scores. Variation in glycan abundances and distribution was most pronounced between different strains of virus (similarity score = 383 out of 999), whereas digestion replicates and injection replicates showed relatively little variation (similarity score = 957). Notably, glycan distributions for homologous regions of influenza glycoprotein variants showed low variability. Due to the multiple possible sources of variation and inherent analytical difficulties in site-specific glycan determinations, variations were individually examined for multiple factors, including differences in supplier, production batch, protease digestion, and replicate measurement. After comparing all glycosylation distributions, four distinguishable classes could be identified for the majority of sites. Finally, attempts to identify glycosylation distributions on adjacent potential N-glycosylated sites of one HA variant were made. Only the second site (NnST) was found to be occupied using two rarely used proteases in proteomics, subtilisin and esperase, both of which did selectively cleave these adjacent sites.
Collapse
Affiliation(s)
- Zachary C Goecker
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA.
| | - Meghan C Burke
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Concepcion A Remoroza
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Yi Liu
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Yuri A Mirokhin
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Sergey L Sheetlin
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Dmitrii V Tchekhovskoi
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Xiaoyu Yang
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| | - Stephen E Stein
- Mass Spectrometry Data Center, National Institute of Standards and Technology, Gaithersburg, Maryland, USA
| |
Collapse
|
25
|
Klemanska A, Dwyer K, Walsh G. Truncation of a novel C-terminal domain of a β-glucanase improves its thermal stability and specific activity. Biotechnol J 2024; 19:e2400245. [PMID: 39118577 DOI: 10.1002/biot.202400245] [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: 04/12/2024] [Revised: 06/10/2024] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Enzymes that degrade β-glucan play important roles in various industries, including those related to brewing, animal feed, and health care. Csph16A, an endo-β-1,3(4)-glucanase encoded by a gene from the halotolerant, xerotolerant, and radiotrophic black fungus Cladosporium sphaerospermum, was cloned and expressed in Pichia pastoris. Two isoforms (Csph16A.1 and Csph16A.2) are produced, arising from differential glycosylation. The proteins were predicted to contain a catalytic Lam16A domain, along with a C-terminal domain (CTD) of unknown function which exhibits minimal secondary structure. Employing PCR-mediated gene truncation, the CTD of Csph16A was excised to assess its functional impact on the enzyme and determine potential alterations in biotechnologically relevant characteristics. The truncated mutant, Csph16A-ΔC, exhibited significantly enhanced thermal stability at 50°C, with D-values 14.8 and 23.5 times greater than those of Csph16A.1 and Csph16A.2, respectively. Moreover, Csph16A-ΔC demonstrated a 20%-25% increase in halotolerance at 1.25 and 1.5 M NaCl, respectively, compared to the full-length enzymes. Notably, specific activity against cereal β-glucan, lichenan, and curdlan was increased by up to 238%. This study represents the first characterization of a glucanase from the stress-tolerant fungus C. sphaerospermum and the first report of a halotolerant and engineered endo-β-1,3(4)-glucanase. Additionally, it sheds light on a group of endo-β-1,3(4)-glucanases from Antarctic rock-inhabiting black fungi harboring a Lam16A catalytic domain and a novel CTD of unknown function.
Collapse
Affiliation(s)
- Anastasia Klemanska
- Department of Chemical Sciences & Bernal Institute, University of Limerick, Castletroy, Limerick City, Ireland
- Monaghan Mushrooms, Tyholland, Co, Monaghan, Ireland
| | - Kelly Dwyer
- Monaghan Mushrooms, Tyholland, Co, Monaghan, Ireland
| | - Gary Walsh
- Department of Chemical Sciences & Bernal Institute, University of Limerick, Castletroy, Limerick City, Ireland
| |
Collapse
|
26
|
Usama, Khan Z, Ali A, Shah M, Imran M. Differential glycosylation in mutant vitamin D-binding protein decimates the binding stability of vitamin D. J Biomol Struct Dyn 2024; 42:5365-5375. [PMID: 37357441 DOI: 10.1080/07391102.2023.2226742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/10/2023] [Indexed: 06/27/2023]
Abstract
Vitamin D (VD) is produced by the skin upon exposure to sunlight or is obtained from dietary sources. Several risk factors are associated with VD deficiency including mutations and post-translational modifications in its transport protein known as vitamin D binding protein (VDBP) or GC-globulin. The two common single nucleotide polymorphisms rs7041 and rs4588 create three major isoforms of VDBP, including GC-1F also called wild type, GC1S, and GC-2. The 3D models for both GC-1F and GC-2 were constructed in their glycosylated states to decipher the effect of these mutations on the overall conformational changes and VD-binding affinity. The binding affinities were estimated using the Molecular Mechanics Poison-Boltzmann surface area (MM-PBSA) method and conformational changes were investigated after free energy landscapes estimations. Total free energies suggest that GC-1F exhibits stronger affinity (ΔE = -116.09 kJ/mol) than GC-2 (ΔE = -95 kJ/mol) variant with VD. The GC-1F isoforms had more streamlined motion compared to GC-2 isoforms, predicting a trade-off between cross-talk residues that significantly impacts protein structural stability. The data suggest that glycation at Thr418 plays a vital role in the overall VDBP-VD affinity by stabilizing the N-T loop that holds the domain I (VD-pocket) and domain III intact. The loss of glycation in GC-2 has a pivotal role in the inter-domain conformational stability of VDBP, which may ultimately affect VD transportation and maturation. These findings describe a novel mechanism in how mutations distant from the VD-active site change the overall conformational of the VDBP and abrogate the VDBP-VD interaction.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Usama
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Zahid Khan
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Aktar Ali
- Biological Screening Core, Warren Family Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana, USA
| | - Masaud Shah
- Department of Physiology, School of Medicine, Ajou University, Suwon, South Korea
| | - Muhammad Imran
- Biochemistry Section, Institute of Chemical Sciences, University of Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
27
|
Wang Y, Liu Y, Liu S, Cheng L, Liu X. Recent advances in N-glycan biomarker discovery among human diseases. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1156-1171. [PMID: 38910518 PMCID: PMC11464920 DOI: 10.3724/abbs.2024101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
N-glycans play important roles in a variety of biological processes. In recent years, analytical technologies with high resolution and sensitivity have advanced exponentially, enabling analysts to investigate N-glycomic changes in different states. Specific glycan and glycosylation signatures have been identified in multiple diseases, including cancer, autoimmune diseases, nervous system disorders, and metabolic and cardiovascular diseases. These glycans demonstrate comparable or superior indicating capability in disease diagnosis and prognosis over routine biomarkers. Moreover, synchronous glycan alterations concurrent with disease initiation and progression provide novel insights into pathogenetic mechanisms and potential treatment targets. This review elucidates the biological significance of N-glycans, compares the existing glycomic technologies, and delineates the clinical performance of N-glycans across a range of diseases.
Collapse
Affiliation(s)
- Yi Wang
- Department of Laboratory MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Yuanyuan Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key LaboratorySystems Biology ThemeDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| | - Si Liu
- Department of Epidemiology and Health StatisticsSchool of Public HealthFujian Medical UniversityFuzhou350122China
| | - Liming Cheng
- Department of Laboratory MedicineTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Xin Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key LaboratorySystems Biology ThemeDepartment of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| |
Collapse
|
28
|
Garmeh Motlagh F, Azimzadeh Irani M, Masoomi Nomandan SZ, Assadizadeh M. Computational design and investigation of the monomeric spike SARS-CoV-2-ferritin nanocage vaccine stability and interactions. Front Mol Biosci 2024; 11:1403635. [PMID: 38933369 PMCID: PMC11199398 DOI: 10.3389/fmolb.2024.1403635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Since the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) outbreak, several solutions have been proposed to manage the disease. The most viable option for controlling this virus is to produce effective vaccines. Most of the current SARS-CoV-2 vaccines have focused on the infusion spike protein. Spike exists as a trimer and plays a vital role in infecting host cells by binding to the Angiotensin-Converting Enzyme 2 (ACE2) receptor through its Receptor Binding Domain (RBD). Ferritin protein, a naturally occurring iron-storage protein, has gained attention for vaccine production due to its self-assembling property, non-toxic nature, and biocompatibility. Ferritin nanocages have recently been employed in the development of a SARS-CoV-2 vaccination eliciting not only long-term protective memory cells but also a sustained antibody response. In this study, a combination of in silico investigations including molecular docking, molecular dynamics simulations, and immune simulations were carried out to computationally model the monomeric spike protein on the ferritin nanocage as well as to evaluate its stability and interactions for the first time. The structural dynamics of the modeled complex demonstrated noticeable stability. In particular, the Receptor Binding Domain (RBD) and ferritin within the monomeric spike-ferritin complex illustrated significant stability. The lack of alterations in the secondary structure further supported the overall steadiness of the complex. The decline in the distance between ferritin and spike suggests a strong interaction over time. The cross-correlation matrices revealed that the monomeric spike and ferritin move towards each other supporting the stable interaction between spike and ferritin. Further, the orientation of monomeric spike protein within the ferritin unit facilitated the exposure of critical epitopes, specifically upward active Receptor Binding Domain (RBD), enabling effective interactions with the ACE2 receptor. The immune simulations of the model indicated high-level stimulations of both cellular and humoral immunity in the human body. It was also found that the employed model is effective regardless of the mutated spikes in different variants. These findings shed light on the current status of the SARS-CoV-2-ferritin nanoparticle vaccines and could be used as a framework for other similar vaccine designs.
Collapse
|
29
|
Whittington C, Sharma A, Hill SG, Iavarone AT, Hoffman BM, Offenbacher AR. Impact of N-Glycosylation on Protein Structure and Dynamics Linked to Enzymatic C-H Activation in the M. oryzae Lipoxygenase. Biochemistry 2024; 63:1335-1346. [PMID: 38690768 PMCID: PMC11587536 DOI: 10.1021/acs.biochem.4c00109] [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] [Indexed: 05/03/2024]
Abstract
Lipoxygenases (LOXs) from pathogenic fungi are potential therapeutic targets for defense against plant and select human diseases. In contrast to the canonical LOXs in plants and animals, fungal LOXs are unique in having appended N-linked glycans. Such important post-translational modifications (PTMs) endow proteins with altered structure, stability, and/or function. In this study, we present the structural and functional outcomes of removing or altering these surface carbohydrates on the LOX from the devastating rice blast fungus, M. oryzae, MoLOX. Alteration of the PTMs did notinfluence the active site enzyme-substrate ground state structures as visualized by electron-nuclear double resonance (ENDOR) spectroscopy. However, removal of the eight N-linked glycans by asparagine-to-glutamine mutagenesis nonetheless led to a change in substrate selectivity and an elevated activation energy for the reaction with substrate linoleic acid, as determined by kinetic measurements. Comparative hydrogen-deuterium exchange mass spectrometry (HDX-MS) analysis of wild-type and Asn-to-Gln MoLOX variants revealed a regionally defined impact on the dynamics of the arched helix that covers the active site. Guided by these HDX results, a single glycan sequon knockout was generated at position 72, and its comparative substrate selectivity from kinetics nearly matched that of the Asn-to-Gln variant. The cumulative data from model glyco-enzyme MoLOX showcase how the presence, alteration, or removal of even a single N-linked glycan can influence the structural integrity and dynamics of the protein that are linked to an enzyme's catalytic proficiency, while indicating that extensive glycosylation protects the enzyme during pathogenesis by protecting it from protease degradation.
Collapse
Affiliation(s)
- Chris Whittington
- Department of Chemistry, East Carolina University, Greenville NC, 27858, United States
| | - Ajay Sharma
- Department of Chemistry, Northwestern University, Evanston, IL 60208, United States
| | - S. Gage Hill
- Department of Chemistry, East Carolina University, Greenville NC, 27858, United States
| | - Anthony T. Iavarone
- California Institute for Quantitative Biosciences, Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, United States
| | - Brian M. Hoffman
- Department of Chemistry, Northwestern University, Evanston, IL 60208, United States
| | - Adam R. Offenbacher
- Department of Chemistry, East Carolina University, Greenville NC, 27858, United States
| |
Collapse
|
30
|
Rowland RR, Brandariz-Nuñez A. Role of N-linked glycosylation in porcine reproductive and respiratory syndrome virus (PRRSV) infection. J Gen Virol 2024; 105:001994. [PMID: 38776134 PMCID: PMC11165596 DOI: 10.1099/jgv.0.001994] [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: 02/28/2024] [Accepted: 05/14/2024] [Indexed: 05/24/2024] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRSV) is an enveloped single-stranded positive-sense RNA virus and one of the main pathogens that causes the most significant economical losses in the swine-producing countries. PRRSV is currently divided into two distinct species, PRRSV-1 and PRRSV-2. The PRRSV virion envelope is composed of four glycosylated membrane proteins and three non-glycosylated envelope proteins. Previous work has suggested that PRRSV-linked glycans are critical structural components for virus assembly. In addition, it has been proposed that PRRSV glycans are implicated in the interaction with host cells and critical for virus infection. In contrast, recent findings showed that removal of N-glycans from PRRSV does not influence virus infection of permissive cells. Thus, there are not sufficient evidences to indicate compellingly that N-glycans present in the PRRSV envelope play a direct function in viral infection. To gain insights into the role of N-glycosylation in PRRSV infection, we analysed the specific contribution of the envelope protein-linked N-glycans to infection of permissive cells. For this purpose, we used a novel strategy to modify envelope protein-linked N-glycans that consists of production of monoglycosylated PRRSV and viral glycoproteins with different glycan states. Our results showed that removal or alteration of N-glycans from PRRSV affected virus infection. Specifically, we found that complex N-glycans are required for an efficient infection in cell cultures. Furthermore, we found that presence of high mannose type glycans on PRRSV surface is the minimal requirement for a productive viral infection. Our findings also show that PRRSV-1 and PRRSV-2 have different requirements of N-glycan structure for an optimal infection. In addition, we demonstrated that removal of N-glycans from PRRSV does not affect viral attachment, suggesting that these carbohydrates played a major role in regulating viral entry. In agreement with these findings, by performing immunoprecipitation assays and colocalization experiments, we found that N-glycans present in the viral envelope glycoproteins are not required to bind to the essential viral receptor CD163. Finally, we found that the presence of N-glycans in CD163 is not required for PRRSV infection.
Collapse
Affiliation(s)
- Raymond R.R. Rowland
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - Alberto Brandariz-Nuñez
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| |
Collapse
|
31
|
Zhao J, Sun H, Wang C, Shang D. Breast cancer therapy: from the perspective of glucose metabolism and glycosylation. Mol Biol Rep 2024; 51:546. [PMID: 38642246 DOI: 10.1007/s11033-024-09466-w] [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: 02/12/2024] [Accepted: 03/22/2024] [Indexed: 04/22/2024]
Abstract
Breast cancer is a leading cause of mortality and the most prevalent form of malignant tumor among women worldwide. Breast cancer cells exhibit an elevated glycolysis and altered glucose metabolism. Moreover, these cells display abnormal glycosylation patterns, influencing invasion, proliferation, metastasis, and drug resistance. Consequently, targeting glycolysis and mitigating abnormal glycosylation represent key therapeutic strategies for breast cancer. This review underscores the importance of protein glycosylation and glucose metabolism alterations in breast cancer. The current research efforts in developing effective interventions targeting glycolysis and glycosylation are further discussed.
Collapse
Affiliation(s)
- Jiaqi Zhao
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Haiting Sun
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China
| | - Che Wang
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian, 116029, China.
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Science, Liaoning Normal University, Dalian, 116081, China.
| | - Dejing Shang
- Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, School of Life Science, Liaoning Normal University, Dalian, 116081, China.
| |
Collapse
|
32
|
Fujitani N, Uehara Y, Ariki S, Hashimoto U, Mukai J, Hasegawa Y, Takahashi M. Site-specific glycosylation analysis of epidermal growth factor receptor 2 (ErbB2): exploring structure and function toward therapeutic targeting. Glycobiology 2024; 34:cwad100. [PMID: 38109791 PMCID: PMC10987295 DOI: 10.1093/glycob/cwad100] [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: 07/30/2023] [Revised: 11/24/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023] Open
Abstract
Glycans found on receptor tyrosine kinases (RTKs) have emerged as promising targets for cancer chemotherapy, aiming to address issues such as drug resistance. However, to effectively select the target glycans, it is crucial to define the structure and function of candidate glycans in advance. Through mass spectrometric analysis, this study presents a "glycoform atlas" of epidermal growth factor receptor 2 (ErbB2), an RTK targeted for the treatment of ErbB2-positive cancers. Our analysis provides an in-depth and site-specific glycosylation profile, including both asparagine- and serine/threonine-linked glycosylation. Molecular dynamics simulations of N-glycosylated ErbB2 incorporating the identified glycan structures suggested that the N-glycan at N124 on the long flexible loop in the N-terminal region plays a role in stabilizing the ErbB2 structure. Based on the model structures obtained from the simulations, analysis employing an ErbB2 mutant deficient in N-glycosylation at N124 exhibited a significantly shorter intracellular half-life and suppressed autophosphorylation compared to wild-type ErbB2. Moreover, a structural comparison between the N-glycosylated forms of ErbB2 and its structurally homologous receptor, epidermal growth factor receptor (EGFR), demonstrated distinct variations in the distribution and density of N-glycans across these two molecules. These findings provide valuable insights into the structural and functional implications of ErbB2 glycosylation and will contribute to facilitating the establishment of glycan-targeted therapeutic strategies for ErbB2-positive cancers.
Collapse
Affiliation(s)
- Naoki Fujitani
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Yasuaki Uehara
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Shigeru Ariki
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
- Department of Chemistry, Sapporo Medical University Center for Medical Education, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Ukichiro Hashimoto
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Jo Mukai
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Yoshihiro Hasegawa
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| | - Motoko Takahashi
- Department of Biochemistry, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo 060-8556, Japan
| |
Collapse
|
33
|
Liu HZ, Song XQ, Zhang H. Sugar-coated bullets: Unveiling the enigmatic mystery 'sweet arsenal' in osteoarthritis. Heliyon 2024; 10:e27624. [PMID: 38496870 PMCID: PMC10944269 DOI: 10.1016/j.heliyon.2024.e27624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
Glycosylation is a crucial post-translational modification process where sugar molecules (glycans) are covalently linked to proteins, lipids, or other biomolecules. In this highly regulated and complex process, a series of enzymes are involved in adding, modifying, or removing sugar residues. This process plays a pivotal role in various biological functions, influencing the structure, stability, and functionality of the modified molecules. Glycosylation is essential in numerous biological processes, including cell adhesion, signal transduction, immune response, and biomolecular recognition. Dysregulation of glycosylation is associated with various diseases. Glycation, a post-translational modification characterized by the non-enzymatic attachment of sugar molecules to proteins, has also emerged as a crucial factor in various diseases. This review comprehensively explores the multifaceted role of glycation in disease pathogenesis, with a specific focus on its implications in osteoarthritis (OA). Glycosylation and glycation alterations wield a profound influence on OA pathogenesis, intertwining with disease onset and progression. Diverse studies underscore the multifaceted role of aberrant glycosylation in OA, particularly emphasizing its intricate relationship with joint tissue degradation and inflammatory cascades. Distinct glycosylation patterns, including N-glycans and O-glycans, showcase correlations with inflammatory cytokines, matrix metalloproteinases, and cellular senescence pathways, amplifying the degenerative processes within cartilage. Furthermore, the impact of advanced glycation end-products (AGEs) formation in OA pathophysiology unveils critical insights into glycosylation-driven chondrocyte behavior and extracellular matrix remodeling. These findings illuminate potential therapeutic targets and diagnostic markers, signaling a promising avenue for targeted interventions in OA management. In this comprehensive review, we aim to thoroughly examine the significant impact of glycosylation or AGEs in OA and explore its varied effects on other related conditions, such as liver-related diseases, immune system disorders, and cancers, among others. By emphasizing glycosylation's role beyond OA and its implications in other diseases, we uncover insights that extend beyond the immediate focus on OA, potentially revealing novel perspectives for diagnosing and treating OA.
Collapse
Affiliation(s)
- Hong-zhi Liu
- Department of Orthopaedics, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin-qiu Song
- The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Hongmei Zhang
- Department of Orthopaedics, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
34
|
SureshKumar H, Appadurai R, Srivastava A. Glycans modulate lipid binding in Lili-Mip lipocalin protein: insights from molecular simulations and protein network analyses. Glycobiology 2024; 34:cwad094. [PMID: 38015986 DOI: 10.1093/glycob/cwad094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 11/30/2023] Open
Abstract
The unique viviparous Pacific Beetle cockroaches provide nutrition to their embryo by secreting milk proteins Lili-Mip, a lipid-binding glycoprotein that crystallises in-vivo. The resolved in-vivo crystal structure of variably glycosylated Lili-Mip shows a classical Lipocalin fold with an eight-stranded antiparallel beta-barrel enclosing a fatty acid. The availability of physiologically unaltered glycoprotein structure makes Lili-Mip a very attractive model system to investigate the role of glycans on protein structure, dynamics, and function. Towards that end, we have employed all-atom molecular dynamics simulations on various glycosylated stages of a bound and free Lili-Mip protein and characterised the impact of glycans and the bound lipid on the dynamics of this glycoconjugate. Our work provides important molecular-level mechanistic insights into the role of glycans in the nutrient storage function of the Lili-Mip protein. Our analyses show that the glycans stabilise spatially proximal residues and regulate the low amplitude opening motions of the residues at the entrance of the binding pocket. Glycans also preserve the native orientation and conformational flexibility of the ligand. However, we find that either deglycosylation or glycosylation with high-mannose and paucimannose on the core glycans, which better mimic the natural insect glycosylation state, significantly affects the conformation and dynamics. A simple but effective distance- and correlation-based network analysis of the protein also reveals the key residues regulating the barrel's architecture and ligand binding characteristics in response to glycosylation.
Collapse
Affiliation(s)
- Harini SureshKumar
- Molecular Biophysics Unit, Indian Institute of Science, C. V. Raman Road, Bangalore, KA 560012, India
| | - Rajeswari Appadurai
- Molecular Biophysics Unit, Indian Institute of Science, C. V. Raman Road, Bangalore, KA 560012, India
| | - Anand Srivastava
- Molecular Biophysics Unit, Indian Institute of Science, C. V. Raman Road, Bangalore, KA 560012, India
| |
Collapse
|
35
|
Pegg CL, Modhiran N, Parry RH, Liang B, Amarilla AA, Khromykh AA, Burr L, Young PR, Chappell K, Schulz BL, Watterson D. The role of N-glycosylation in spike antigenicity for the SARS-CoV-2 gamma variant. Glycobiology 2024; 34:cwad097. [PMID: 38048640 PMCID: PMC10969516 DOI: 10.1093/glycob/cwad097] [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: 06/28/2023] [Revised: 11/10/2023] [Accepted: 11/28/2023] [Indexed: 12/06/2023] Open
Abstract
The emergence of SARS-CoV-2 variants alters the efficacy of existing immunity towards the viral spike protein, whether acquired from infection or vaccination. Mutations that impact N-glycosylation of spike may be particularly important in influencing antigenicity, but their consequences are difficult to predict. Here, we compare the glycosylation profiles and antigenicity of recombinant viral spike of ancestral Wu-1 and the Gamma strain, which has two additional N-glycosylation sites due to amino acid substitutions in the N-terminal domain (NTD). We found that a mutation at residue 20 from threonine to asparagine within the NTD caused the loss of NTD-specific antibody COVA2-17 binding. Glycan site-occupancy analyses revealed that the mutation resulted in N-glycosylation switching to the new sequon at N20 from the native N17 site. Site-specific glycosylation profiles demonstrated distinct glycoform differences between Wu-1, Gamma, and selected NTD variant spike proteins, but these did not affect antibody binding. Finally, we evaluated the specificity of spike proteins against convalescent COVID-19 sera and found reduced cross-reactivity against some mutants, but not Gamma spike compared to Wuhan spike. Our results illustrate the impact of viral divergence on spike glycosylation and SARS-CoV-2 antibody binding profiles.
Collapse
Affiliation(s)
- Cassandra L Pegg
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, Building 75, Corner College Road and Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Rhys H Parry
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Benjamin Liang
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Alexander A Khromykh
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| | - Lucy Burr
- Department of Respiratory Medicine, Mater Health Services, Raymond Terrace, South Brisbane, Queensland 4101, Australia
| | - Paul R Young
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, Building 75, Corner College Road and Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| | - Keith Chappell
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, Building 75, Corner College Road and Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| | - Benjamin L Schulz
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, Building 75, Corner College Road and Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| |
Collapse
|
36
|
Hesamzadeh P, Seif A, Mahmoudzadeh K, Ganjali Koli M, Mostafazadeh A, Nayeri K, Mirjafary Z, Saeidian H. De novo antioxidant peptide design via machine learning and DFT studies. Sci Rep 2024; 14:6473. [PMID: 38499731 PMCID: PMC10948870 DOI: 10.1038/s41598-024-57247-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/15/2024] [Indexed: 03/20/2024] Open
Abstract
Antioxidant peptides (AOPs) are highly valued in food and pharmaceutical industries due to their significant role in human function. This study introduces a novel approach to identifying robust AOPs using a deep generative model based on sequence representation. Through filtration with a deep-learning classification model and subsequent clustering via the Butina cluster algorithm, twelve peptides (GP1-GP12) with potential antioxidant capacity were predicted. Density functional theory (DFT) calculations guided the selection of six peptides for synthesis and biological experiments. Molecular orbital representations revealed that the HOMO for these peptides is primarily localized on the indole segment, underscoring its pivotal role in antioxidant activity. All six synthesized peptides exhibited antioxidant activity in the DPPH assay, while the hydroxyl radical test showed suboptimal results. A hemolysis assay confirmed the non-hemolytic nature of the generated peptides. Additionally, an in silico investigation explored the potential inhibitory interaction between the peptides and the Keap1 protein. Analysis revealed that ligands GP3, GP4, and GP12 induced significant structural changes in proteins, affecting their stability and flexibility. These findings highlight the capability of machine learning approaches in generating novel antioxidant peptides.
Collapse
Affiliation(s)
- Parsa Hesamzadeh
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Abdolvahab Seif
- Dipartimento di Fisica, Universita' di Padova, Via Marzolo 8, 35131, Padua, Italy
- Department of Chemistry, University of Turin, Via Pietro Giuria 7, 10125, Turin, Italy
| | - Kazem Mahmoudzadeh
- Department of Organic Chemistry and Oil, Faculty of Chemistry, Shahid Beheshti University, Tehran, Iran
| | | | - Amrollah Mostafazadeh
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Kosar Nayeri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Zohreh Mirjafary
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hamid Saeidian
- Department of Science, Payame Noor University (PNU), PO Box: 19395-4697, Tehran, Iran.
| |
Collapse
|
37
|
Drzewicka K, Zasłona Z. Metabolism-driven glycosylation represents therapeutic opportunities in interstitial lung diseases. Front Immunol 2024; 15:1328781. [PMID: 38550597 PMCID: PMC10973144 DOI: 10.3389/fimmu.2024.1328781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024] Open
Abstract
Metabolic changes are coupled with alteration in protein glycosylation. In this review, we will focus on macrophages that are pivotal in the pathogenesis of pulmonary fibrosis and sarcoidosis and thanks to their adaptable metabolism are an attractive therapeutic target. Examples presented in this review demonstrate that protein glycosylation regulates metabolism-driven immune responses in macrophages, with implications for fibrotic processes and granuloma formation. Targeting proteins that regulate glycosylation, such as fucosyltransferases, neuraminidase 1 and chitinase 1 could effectively block immunometabolic changes driving inflammation and fibrosis, providing novel avenues for therapeutic interventions.
Collapse
|
38
|
Pasala C, Sharma S, Roychowdhury T, Moroni E, Colombo G, Chiosis G. N-Glycosylation as a Modulator of Protein Conformation and Assembly in Disease. Biomolecules 2024; 14:282. [PMID: 38540703 PMCID: PMC10968129 DOI: 10.3390/biom14030282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 05/01/2024] Open
Abstract
Glycosylation, a prevalent post-translational modification, plays a pivotal role in regulating intricate cellular processes by covalently attaching glycans to macromolecules. Dysregulated glycosylation is linked to a spectrum of diseases, encompassing cancer, neurodegenerative disorders, congenital disorders, infections, and inflammation. This review delves into the intricate interplay between glycosylation and protein conformation, with a specific focus on the profound impact of N-glycans on the selection of distinct protein conformations characterized by distinct interactomes-namely, protein assemblies-under normal and pathological conditions across various diseases. We begin by examining the spike protein of the SARS virus, illustrating how N-glycans regulate the infectivity of pathogenic agents. Subsequently, we utilize the prion protein and the chaperone glucose-regulated protein 94 as examples, exploring instances where N-glycosylation transforms physiological protein structures into disease-associated forms. Unraveling these connections provides valuable insights into potential therapeutic avenues and a deeper comprehension of the molecular intricacies that underlie disease conditions. This exploration of glycosylation's influence on protein conformation effectively bridges the gap between the glycome and disease, offering a comprehensive perspective on the therapeutic implications of targeting conformational mutants and their pathologic assemblies in various diseases. The goal is to unravel the nuances of these post-translational modifications, shedding light on how they contribute to the intricate interplay between protein conformation, assembly, and disease.
Collapse
Affiliation(s)
- Chiranjeevi Pasala
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.P.); (S.S.); (T.R.)
| | - Sahil Sharma
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.P.); (S.S.); (T.R.)
| | - Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.P.); (S.S.); (T.R.)
| | - Elisabetta Moroni
- The Institute of Chemical Sciences and Technologies (SCITEC), Italian National Research Council (CNR), 20131 Milano, Italy; (E.M.); (G.C.)
| | - Giorgio Colombo
- The Institute of Chemical Sciences and Technologies (SCITEC), Italian National Research Council (CNR), 20131 Milano, Italy; (E.M.); (G.C.)
- Department of Chemistry, University of Pavia, 27100 Pavia, Italy
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.P.); (S.S.); (T.R.)
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
39
|
Poddar S, Roy R, Kar P. The conformational dynamics of Hepatitis C Virus E2 glycoprotein with the increasing number of N-glycosylation unraveled by molecular dynamics simulations. J Biomol Struct Dyn 2024:1-16. [PMID: 38393644 DOI: 10.1080/07391102.2024.2319679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 02/12/2024] [Indexed: 02/25/2024]
Abstract
The Hepatitis C Virus (HCV), responsible for causing hepatitis and a significant contributor to liver disorders, presents a challenge for treatment due to its high genetic variability. Despite efforts, there is still no effective medication available for this virus. One of the promising targets for drug development involves targeting glycoprotein E2. However, our understanding of the dynamic behavior of E2 and its associated glycans remains limited. In this study, we investigated the dynamic characteristics of E2 with varying degrees of glycosylation using all-atom molecular dynamics simulations. We also explored glycan's interactions with the protein and among themselves. An overall increase in correlation between the vital protein regions was observed with an increase in glycan number. The protein dynamics is followed by the analysis of glycan dynamics, where the flexibility of the individual glycans was analyzed in their free and bound state, which revealed a decrease in their fluctuation in some cases. Furthermore, we generated the free energy landscape of individual N-glycan linkages in both free and bound states and observed both increases and decreases in flexibility, which can be attributed to the formation and breakage of hydrogen bonds with amino acids. Finally, we found that for a high glycosylation system, glycans interact with glycoprotein and form hydrogen bonds among themselves. Moreover, the hydrogen bond profiles of a given glycan can vary when influenced by other glycans. In summary, our study provides valuable insights into the dynamics of the core region of HCV E2 glycoprotein and its associated glycans.
Collapse
Affiliation(s)
- Sayan Poddar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, India
| | - Rajarshi Roy
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, India
| |
Collapse
|
40
|
Kosutova N, Lorencova L, Hires M, Jane E, Orovcik L, Kollar J, Kozics K, Gabelova A, Ukraintsev E, Rezek B, Kasak P, Cernocka H, Ostatna V, Blahutova J, Vikartovska A, Bertok T, Tkac J. Negative Charge-Carrying Glycans Attached to Exosomes as Novel Liquid Biopsy Marker. SENSORS (BASEL, SWITZERLAND) 2024; 24:1128. [PMID: 38400284 PMCID: PMC10892626 DOI: 10.3390/s24041128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Abstract
Prostate cancer (PCa) is the second most common cancer. In this paper, the isolation and properties of exosomes as potential novel liquid biopsy markers for early PCa liquid biopsy diagnosis are investigated using two prostate human cell lines, i.e., benign (control) cell line RWPE1 and carcinoma cell line 22Rv1. Exosomes produced by both cell lines are characterised by various methods including nanoparticle-tracking analysis, dynamic light scattering, scanning electron microscopy and atomic force microscopy. In addition, surface plasmon resonance (SPR) is used to study three different receptors on the exosomal surface (CD63, CD81 and prostate-specific membrane antigen-PMSA), implementing monoclonal antibodies and identifying the type of glycans present on the surface of exosomes using lectins (glycan-recognising proteins). Electrochemical analysis is used to understand the interfacial properties of exosomes. The results indicate that cancerous exosomes are smaller, are produced at higher concentrations, and exhibit more nega tive zeta potential than the control exosomes. The SPR experiments confirm that negatively charged α-2,3- and α-2,6-sialic acid-containing glycans are found in greater abundance on carcinoma exosomes, whereas bisecting and branched glycans are more abundant in the control exosomes. The SPR results also show that a sandwich antibody/exosomes/lectins configuration could be constructed for effective glycoprofiling of exosomes as a novel liquid biopsy marker.
Collapse
Affiliation(s)
- Natalia Kosutova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Lenka Lorencova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Michal Hires
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Eduard Jane
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Lubomir Orovcik
- Institute of Materials and Machine Mechanics, Slovak Academy of Sciences, Dubravska cesta 9/6319, 845 13 Bratislava, Slovakia
| | - Jozef Kollar
- Polymer Institute, Slovak Academy of Sciences, Dubravska cesta 9, 845 41 Bratislava, Slovakia
| | - Katarina Kozics
- Biomedical Research Centre, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia (A.G.)
| | - Alena Gabelova
- Biomedical Research Centre, Slovak Academy of Sciences, Dubravska cesta 9, 845 05 Bratislava, Slovakia (A.G.)
| | - Egor Ukraintsev
- Department of Physics, Faculty of Electrical Engineering, Czech Technical University in Prague, Technicka 2, 166 27 Prague, Czech Republic; (E.U.); (B.R.)
| | - Bohuslav Rezek
- Department of Physics, Faculty of Electrical Engineering, Czech Technical University in Prague, Technicka 2, 166 27 Prague, Czech Republic; (E.U.); (B.R.)
| | - Peter Kasak
- Centre for Advanced Materials, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Hana Cernocka
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, 61200 Brno, Czech Republic; (H.C.)
| | - Veronika Ostatna
- Institute of Biophysics, Czech Academy of Sciences, Kralovopolska 135, 61200 Brno, Czech Republic; (H.C.)
| | - Jana Blahutova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Alica Vikartovska
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Tomas Bertok
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| | - Jan Tkac
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 5807/9, 845 38 Bratislava, Slovakia (L.L.); (E.J.)
| |
Collapse
|
41
|
Dixit B, Vranken W, Ghysels A. Conformational dynamics of α-1 acid glycoprotein (AGP) in cancer: A comparative study of glycosylated and unglycosylated AGP. Proteins 2024; 92:246-264. [PMID: 37837263 DOI: 10.1002/prot.26607] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/01/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
α-1 acid glycoprotein (AGP) is one of the most abundant plasma proteins. It fulfills two important functions: immunomodulation, and binding to various drugs and receptors. These different functions are closely associated and modulated via changes in glycosylation and cancer missense mutations. From a structural point of view, glycans alter the local biophysical properties of the protein leading to a diverse ligand-binding spectrum. However, glycans can typically not be observed in the resolved X-ray crystallography structure of AGP due to their high flexibility and microheterogeneity, so limiting our understanding of AGP's conformational dynamics 70 years after its discovery. We here investigate how mutations and glycosylation interfere with AGP's conformational dynamics changing its biophysical behavior, by using molecular dynamics (MD) simulations and sequence-based dynamics predictions. The MD trajectories show that glycosylation decreases the local backbone flexibility of AGP and increases the flexibility of distant regions through allosteric effects. We observe that mutations near the glycosylation site affect glycan's conformational preferences. Thus, we conclude that mutations control glycan dynamics which modulates the protein's backbone flexibility directly affecting its accessibility. These findings may assist in the drug design targeting AGP's glycosylation and mutations in cancer.
Collapse
Affiliation(s)
- Bhawna Dixit
- IBiTech-BioMMeda Group, Ghent University, Ghent, Belgium
- Interuniversity Institute of Bioinformatics in Brussels, ULB-VUB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wim Vranken
- Interuniversity Institute of Bioinformatics in Brussels, ULB-VUB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - An Ghysels
- IBiTech-BioMMeda Group, Ghent University, Ghent, Belgium
| |
Collapse
|
42
|
Wahhab BH, Oyewusi HA, Wahab RA, Mohammad Hood MH, Abdul Hamid AA, Al-Nimer MS, Edbeib MF, Kaya Y, Huyop F. Comparative modeling and enzymatic affinity of novel haloacid dehalogenase from Bacillus megaterium strain BHS1 isolated from alkaline Blue Lake in Turkey. J Biomol Struct Dyn 2024; 42:1429-1442. [PMID: 37038649 DOI: 10.1080/07391102.2023.2199870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 04/01/2023] [Indexed: 04/12/2023]
Abstract
This study presents the initial structural model of L-haloacid dehalogenase (DehLBHS1) from Bacillus megaterium BHS1, an alkalotolerant bacterium known for its ability to degrade halogenated environmental pollutants. The model provides insights into the structural features of DehLBHS1 and expands our understanding of the enzymatic mechanisms involved in the degradation of these hazardous pollutants. Key amino acid residues (Arg40, Phe59, Asn118, Asn176, and Trp178) in DehLBHS1 were identified to play critical roles in catalysis and molecular recognition of haloalkanoic acid, essential for efficient binding and transformation of haloalkanoic acid molecules. DehLBHS1 was modeled using I-TASSER, yielding a best TM-score of 0.986 and an RMSD of 0.53 Å. Validation of the model using PROCHECK revealed that 89.2% of the residues were located in the most favored region, providing confidence in its structural accuracy. Molecular docking simulations showed that the non-simulated DehLBHS1 preferred 2,2DCP over other substrates, forming one hydrogen bond with Arg40 and exhibiting a minimum energy of -2.5 kJ/mol. The simulated DehLBHS1 exhibited a minimum energy of -4.3 kJ/mol and formed four hydrogen bonds with Arg40, Asn176, Asp9, and Tyr11, further confirming the preference for 2,2DCP. Molecular dynamics simulations supported this preference, based on various metrics, including RMSD, RMSF, gyration, hydrogen bonding, and molecular distance. MM-PBSA calculations showed that the DehLBHS1-2,2-DCP complex had a markedly lower binding energy (-21.363 ± 1.26 kcal/mol) than the DehLBHS1-3CP complex (-14.327 ± 1.738 kcal/mol). This finding has important implications for the substrate specificity and catalytic function of DehLBHS1, particularly in the bioremediation of 2,2-DCP in contaminated alkaline environments. These results provide a detailed view of the molecular interactions between the enzyme and its substrate and may aid in the development of more efficient biocatalytic strategies for the degradation of halogenated compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Batool Hazim Wahhab
- Department of Microbiology, Faculty of Medicine, Al-Mustansiriyah University, Iraq
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Malaysia
| | - Habeebat Adekilekun Oyewusi
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Malaysia
- Department of Biochemistry, School of Science and Computer Studies, Federal Polytechnic Ado Ekiti, Ekiti State, Nigeria
| | - Roswanira Abdul Wahab
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia, Malaysia
| | - Mohammad Hakim Mohammad Hood
- Department of Biotechnology, Kulliyah of Science, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Azzmer Azzar Abdul Hamid
- Department of Biotechnology, Kulliyah of Science, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| | - Marwan Salih Al-Nimer
- Department of Pharmacology, College of Medicine, University of Diyala, Baqubah, Iraq
| | - Mohamed Faraj Edbeib
- Department of Medical Laboratories, Faculty of Medical Technology, Bani Walid University, Libya
| | - Yilmaz Kaya
- Department of Biology, Faculty of Science, Kyrgyz-Turkish Manas University, Bishkek, Kyrgyzstan
- Department of Agricultural Biotechnology, Faculty of Agriculture, Ondokuz Mayis University, Samsun, Turkey
| | - Fahrul Huyop
- Department of Biosciences, Faculty of Science, Universiti Teknologi Malaysia, Malaysia
| |
Collapse
|
43
|
Cramer DAT, Yin V, Caval T, Franc V, Yu D, Wu G, Lloyd G, Langendorf C, Whisstock JC, Law RHP, Heck AJR. Proteoform-Resolved Profiling of Plasminogen Activation Reveals Novel Abundant Phosphorylation Site and Primary N-Terminal Cleavage Site. Mol Cell Proteomics 2024; 23:100696. [PMID: 38101751 PMCID: PMC10825491 DOI: 10.1016/j.mcpro.2023.100696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 12/17/2023] Open
Abstract
Plasminogen (Plg), the zymogen of plasmin (Plm), is a glycoprotein involved in fibrinolysis and a wide variety of other physiological processes. Plg dysregulation has been implicated in a range of diseases. Classically, human Plg is categorized into two types, supposedly having different functional features, based on the presence (type I) or absence (type II) of a single N-linked glycan. Using high-resolution native mass spectrometry, we uncovered that the proteoform profiles of human Plg (and Plm) are substantially more extensive than this simple binary classification. In samples derived from human plasma, we identified up to 14 distinct proteoforms of Plg, including a novel highly stoichiometric phosphorylation site at Ser339. To elucidate the potential functional effects of these post-translational modifications, we performed proteoform-resolved kinetic analyses of the Plg-to-Plm conversion using several canonical activators. This conversion is thought to involve at least two independent cleavage events: one to remove the N-terminal peptide and another to release the active catalytic site. Our analyses reveal that these processes are not independent but are instead tightly regulated and occur in a step-wise manner. Notably, N-terminal cleavage at the canonical site (Lys77) does not occur directly from intact Plg. Instead, an activation intermediate corresponding to cleavage at Arg68 is initially produced, which only then is further processed to the canonical Lys77 product. Based on our results, we propose a refined categorization for human Plg proteoforms. In addition, we reveal that the proteoform profile of human Plg is more extensive than that of rat Plg, which lacks, for instance, the here-described phosphorylation at Ser339.
Collapse
Affiliation(s)
- Dario A T Cramer
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands
| | - Victor Yin
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands
| | - Tomislav Caval
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands
| | - Vojtech Franc
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands
| | - Dingyi Yu
- Mass Spectrometry Facility, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Guojie Wu
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Gordon Lloyd
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Christopher Langendorf
- Mass Spectrometry Facility, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - James C Whisstock
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Ruby H P Law
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Melbourne, Victoria, Australia.
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Science, University of Utrecht, Utrecht, The Netherlands; Netherlands Proteomics Centre, University of Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
44
|
Senjor E, Pirro M, Švajger U, Prunk M, Sabotič J, Jewett A, Hensbergen PJ, Perišić Nanut M, Kos J. Different glycosylation profiles of cystatin F alter the cytotoxic potential of natural killer cells. Cell Mol Life Sci 2023; 81:8. [PMID: 38092995 PMCID: PMC10719177 DOI: 10.1007/s00018-023-05041-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/13/2023] [Accepted: 11/06/2023] [Indexed: 12/17/2023]
Abstract
Cystatin F, a cysteine peptidase inhibitor, is a potent modulator of NK cytotoxicity. By inhibiting granule-mediated cytotoxicity pathway, cystatin F induces formation of non-functional NK cell stage, called split-anergy. We show that N-glycosylation determines the localization and cellular function of cystatin F. Cystatin F mostly exhibited high-mannose glycosylation in U-937 cells, both high-mannose and complex glycosylation in NK-92 and primary NKs, and predominantly complex glycosylation in super-charged NKs. Manipulating N-glycosylation with kifunensine increased high-mannose glycosylation of cystatin F and lysosome localisation, which decreased cathepsin C activity and reduced NK cytotoxicity. Mannose-6-phosphate could significantly reduce the internalization of extracellular cystatin F. By comparing NK cells with different cytotoxic potentials, we found that high-mannose cystatin F was strongly associated with lysosomes and cathepsin C in NK-92 cell line. In contrast, in highly cytotoxic super-charged NKs, cystatin F with complex glycosylation was associated with the secretory pathway and less prone to inhibit cathepsin C. Modulating glycosylation to alter cystatin F localisation could increase the cytotoxicity of NK cells, thereby enhancing their therapeutic potential for treating cancer patients.
Collapse
Affiliation(s)
- Emanuela Senjor
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia
| | - Martina Pirro
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Urban Švajger
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Mateja Prunk
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Jerica Sabotič
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Anahid Jewett
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, School of Dentistry, University of California Los Angeles, Los Angeles, USA
- The Jonsson Comprehensive Cancer Center, Los Angeles, USA
| | - Paul J Hensbergen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000, Ljubljana, Slovenia.
| |
Collapse
|
45
|
Barnieh FM, Galuska SP, Loadman PM, Ward S, Falconer RA, El-Khamisy SF. Cancer-specific glycosylation of CD13 impacts its detection and activity in preclinical cancer tissues. iScience 2023; 26:108219. [PMID: 37942010 PMCID: PMC10628746 DOI: 10.1016/j.isci.2023.108219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/14/2023] [Accepted: 10/12/2023] [Indexed: 11/10/2023] Open
Abstract
Harnessing the differences between cancer and non-cancer tissues presents new opportunities for selective targeting by anti-cancer drugs. CD13, a heavily glycosylated protein, is one example with significant unmet clinical potential in cancer drug discovery. Despite its high expression and activity in cancers, CD13 is also expressed in many normal tissues. Here, we report differential tissue glycosylation of CD13 across tissues and demonstrate for the first time that the nature and pattern of glycosylation of CD13 in preclinical cancer tissues are distinct compared to normal tissues. We identify cancer-specific O-glycosylation of CD13, which selectively blocks its detection in cancer models but not in normal tissues. In addition, the metabolism activity of cancer-expressed CD13 was observed to be critically dependent on its unique glycosylation. Thus, our data demonstrate the existence of discrete cancer-specific CD13 glycoforms and propose cancer-specific CD13 glycoforms as a clinically useful target for effective cancer-targeted therapy.
Collapse
Affiliation(s)
- Francis M. Barnieh
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Sebastian P. Galuska
- Institute for Reproductive Biology, Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, Dummerstorf, Germany
| | - Paul M. Loadman
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | | | - Robert A. Falconer
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Sherif F. El-Khamisy
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
46
|
Chen H, Zhang Y, Shen Y, Jiang L, Zhang G, Zhang X, Xu Y, Fu F. Deficiency of N-linked glycosylation impairs immune function of B7-H6. Front Immunol 2023; 14:1255667. [PMID: 38035117 PMCID: PMC10684670 DOI: 10.3389/fimmu.2023.1255667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
B7-H6 is a novel immune checkpoint molecule that triggers NK cell cytotoxicity, but the role of N-glycosylation in B7-H6 is poorly understood. We here identified the existence of N-glycosylation of B7-H6 in different cell lines and exogenous expression cells by PNGase F digestion and tunicamycin blockage. Subsequently, we demonstrated that B7-H6 contains 6 functional N-linked glycosylation sites by single site mutation and electrophoresis. Phylogenetical and structural analysis revealed that N43 and N208 glycan are conserved in jawed vertebrates and may thus contribute more to the biological functions. We further demonstrated that N43 and N208 glycosylation are essential for B7-H6 to trigger NK cell activation. Mechanistically, we found that N43 and N208 glycan contributed to the stability and membrane expression of B7-H6 protein. Lack of N208 glycosylation led to membrane B7-H6 shedding, while N43 mutation resulted in impaired B7-H6/NKp30 binding affinity. Together, our findings highlight the significance of N-linked glycosylation in B7-H6 biological functions and suggest potential targets for modulating NK cell-mediated immunity.
Collapse
Affiliation(s)
- Hanqing Chen
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Hematology, the First affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Zhang
- Department of Respiratory and Critical Medicine, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Yu Shen
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liang Jiang
- Suzhou Red Cross Blood Center, Suzhou, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Xu
- Department of Hematology, the First affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fengqing Fu
- Jiangsu Institute of Clinical Immunology, the First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
47
|
Seitz C, Deveci İ, McCammon JA. Glycosylation and Crowded Membrane Effects on Influenza Neuraminidase Stability and Dynamics. J Phys Chem Lett 2023; 14:9926-9934. [PMID: 37903229 PMCID: PMC10641874 DOI: 10.1021/acs.jpclett.3c02524] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/01/2023]
Abstract
All protein simulations are conducted with varying degrees of simplification, oftentimes with unknown ramifications about how these simplifications affect the interpretability of the results. In this work, we investigated how protein glycosylation and lateral crowding effects modulate an array of properties characterizing the stability and dynamics of influenza neuraminidase. We constructed three systems: (1) glycosylated neuraminidase in a whole virion (i.e., crowded membrane) environment, (2) glycosylated neuraminidase in its own lipid bilayer, and (3) unglycosylated neuraminidase in its own lipid bilayer. We saw that glycans tend to stabilize the protein structure and reduce its conformational flexibility while restricting the solvent movement. Conversely, a crowded membrane environment encouraged exploration of the free energy landscape and a large-scale conformational change, while making the protein structure more compact. Understanding these effects informs what factors one must consider in attempting to recapture the desired level of physical accuracy.
Collapse
Affiliation(s)
- Christian Seitz
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - İlker Deveci
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - J. Andrew McCammon
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| |
Collapse
|
48
|
Greisen PJ, Yi L, Zhou R, Zhou J, Johansson E, Dong T, Liu H, Johnsen LB, Lund S, Svensson LA, Zhu H, Thomas N, Yang Z, Østergaard H. Computational design of N-linked glycans for high throughput epitope profiling. Protein Sci 2023; 32:e4726. [PMID: 37421602 PMCID: PMC10521239 DOI: 10.1002/pro.4726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/12/2023] [Accepted: 07/06/2023] [Indexed: 07/10/2023]
Abstract
Efficient identification of epitopes is crucial for drug discovery and design as it enables the selection of optimal epitopes, expansion of lead antibody diversity, and verification of binding interface. Although high-resolution low throughput methods like x-ray crystallography can determine epitopes or protein-protein interactions accurately, they are time-consuming and can only be applied to a limited number of complexes. To overcome these limitations, we have developed a rapid computational method that incorporates N-linked glycans to mask epitopes or protein interaction surfaces, thereby providing a mapping of these regions. Using human coagulation factor IXa (fIXa) as a model system, we computationally screened 158 positions and expressed 98 variants to test experimentally for epitope mapping. We were able to delineate epitopes rapidly and reliably through the insertion of N-linked glycans that efficiently disrupted binding in a site-selective manner. To validate the efficacy of our method, we conducted ELISA experiments and high-throughput yeast surface display assays. Furthermore, x-ray crystallography was employed to verify the results, thereby recapitulating through the method of N-linked glycans a coarse-grained mapping of the epitope.
Collapse
Affiliation(s)
| | - Li Yi
- Global Research TechnologiesNovo Nordisk A/SMaaloevDenmark
| | - Rong Zhou
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | - Jian Zhou
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | - Eva Johansson
- Global Research TechnologiesNovo Nordisk A/SMaaloevDenmark
| | - Tiantang Dong
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | - Haimo Liu
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | | | - Søren Lund
- Global Research TechnologiesNovo Nordisk A/SMaaloevDenmark
| | | | - Haisun Zhu
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | - Nidhin Thomas
- Digital Science and InnovationNovo Nordisk A/SSeattleUSA
| | - Zhiru Yang
- Discovery Technology China, Novo Nordisk Research CentreNovo Nordisk A/SBeijingChina
| | | |
Collapse
|
49
|
Rocamora F, Peralta AG, Shin S, Sorrentino J, Wu MYM, Toth EA, Fuerst TR, Lewis NE. Glycosylation shapes the efficacy and safety of diverse protein, gene and cell therapies. Biotechnol Adv 2023; 67:108206. [PMID: 37354999 PMCID: PMC11168894 DOI: 10.1016/j.biotechadv.2023.108206] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/26/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Over recent decades, therapeutic proteins have had widespread success in treating a myriad of diseases. Glycosylation, a near universal feature of this class of drugs, is a critical quality attribute that significantly influences the physical properties, safety profile and biological activity of therapeutic proteins. Optimizing protein glycosylation, therefore, offers an important avenue to developing more efficacious therapies. In this review, we discuss specific examples of how variations in glycan structure and glycoengineering impacts the stability, safety, and clinical efficacy of protein-based drugs that are already in the market as well as those that are still in preclinical development. We also highlight the impact of glycosylation on next generation biologics such as T cell-based cancer therapy and gene therapy.
Collapse
Affiliation(s)
- Frances Rocamora
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Angelo G Peralta
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Seunghyeon Shin
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - James Sorrentino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mina Ying Min Wu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eric A Toth
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA
| | - Thomas R Fuerst
- Institute for Bioscience and Biotechnology Research, University of Maryland, Rockville, MD 20850, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
50
|
Seitz C, Deveci İ, McCammon JA. Glycosylation and Crowded Membrane Effects on Influenza Neuraminidase Stability and Dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.556910. [PMID: 37745347 PMCID: PMC10515755 DOI: 10.1101/2023.09.10.556910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
All protein simulations are conducted with varying degrees of simplifications, oftentimes with unknown ramifications on how these simplifications affect the interpretability of the results. In this work we investigated how protein glycosylation and lateral crowding effects modulate an array of properties characterizing the stability and dynamics of influenza neuraminidase. We constructed three systems: 1) Glycosylated neuraminidase in a whole virion (i.e. crowded membrane) environment 2) Glycosylated neuraminidase in its own lipid bilayer 3) Unglycosylated neuraminidase in its own lipid bilayer. We saw that glycans tend to stabilize the protein structure and reduce its conformational flexibility while restricting solvent movement. Conversely, a crowded membrane environment encouraged exploration of the free energy landscape and a large scale conformational change while making the protein structure more compact. Understanding these effects informs what factors one must consider while attempting to recapture the desired level of physical accuracy.
Collapse
Affiliation(s)
- Christian Seitz
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California
| | - İlker Deveci
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California
- Department of Pharmacology, University of California, San Diego, La Jolla, California
| |
Collapse
|