1
|
Angulo J, Ardá A, Bertuzzi S, Canales A, Ereño-Orbea J, Gimeno A, Gomez-Redondo M, Muñoz-García JC, Oquist P, Monaco S, Poveda A, Unione L, Jiménez-Barbero J. NMR investigations of glycan conformation, dynamics, and interactions. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2024; 144-145:97-152. [PMID: 39645352 DOI: 10.1016/j.pnmrs.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 12/09/2024]
Abstract
Glycans are ubiquitous in nature, decorating our cells and serving as the initial points of contact with any visiting entities. These glycan interactions are fundamental to host-pathogen recognition and are related to various diseases, including inflammation and cancer. Therefore, understanding the conformations and dynamics of glycans, as well as the key features that regulate their interactions with proteins, is crucial for designing new therapeutics. Due to the intrinsic flexibility of glycans, NMR is an essential tool for unravelling these properties. In this review, we describe the key NMR parameters that can be extracted from the different experiments, and which allow us to deduce the necessary geometry and molecular motion information, with a special emphasis on assessing the internal motions of the glycosidic linkages. We specifically address the NMR peculiarities of various natural glycans, from histo-blood group antigens to glycosaminoglycans, and also consider the special characteristics of their synthetic analogues (glycomimetics). Finally, we discuss the application of NMR protocols to study glycan-related molecular recognition events, both from the carbohydrate and receptor perspectives, including the use of stable isotopes and paramagnetic NMR methods to overcome the inherent degeneracy of glycan chemical shifts.
Collapse
Affiliation(s)
- Jesús Angulo
- Institute for Chemical Research (IIQ), CSIC-University of Seville, 49 Américo Vespucio, 41092 Seville, Spain
| | - Ana Ardá
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Sara Bertuzzi
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Angeles Canales
- Departamento de Química Orgánica, Facultad Ciencias Químicas, Universidad Complutense de Madrid, Avd. Complutense s/n, C.P. 28040 Madrid, Spain
| | - June Ereño-Orbea
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Ana Gimeno
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Marcos Gomez-Redondo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Juan C Muñoz-García
- Institute for Chemical Research (IIQ), CSIC-University of Seville, 49 Américo Vespucio, 41092 Seville, Spain
| | - Paola Oquist
- Departamento de Química Orgánica, Facultad Ciencias Químicas, Universidad Complutense de Madrid, Avd. Complutense s/n, C.P. 28040 Madrid, Spain
| | - Serena Monaco
- School of Pharmacy, University of East Anglia, Norwich Research Park, NR47TJ Norwich, UK
| | - Ana Poveda
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Luca Unione
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain
| | - Jesús Jiménez-Barbero
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Spain; Department of Organic & Inorganic Chemistry, Faculty of Science and Technology, University of the Basque Country, EHU-UPV, 48940 Leioa, Bizkaia, Spain; Centro de Investigacion Biomedica En Red de Enfermedades Respiratorias, 28029 Madrid, Spain.
| |
Collapse
|
2
|
Maier I, Kontaxis G, Zimmermann C, Steininger C. Cyanovirin-N Binding to N-Acetyl-d-glucosamine Requires Carbohydrate-Binding Sites on Two Different Protomers. Biochemistry 2024; 63:1270-1277. [PMID: 38770609 PMCID: PMC11112747 DOI: 10.1021/acs.biochem.4c00113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 05/22/2024]
Abstract
Cyanovirin-N (CV-N) binds high-mannose oligosaccharides on enveloped viruses with two carbohydrate-binding sites, one bearing high affinity and one low affinity to Manα(1-2)Man moieties. A tandem repeat of two CV-N molecules (CVN2) was tested for antiviral activity against human immunodeficiency virus type I (HIV-1) by using a domain-swapped dimer. CV-N was shown to bind N-acetylmannosamine (ManNAc) and N-acetyl-d-glucosamine (GlcNAc) when the carbohydrate-binding sites in CV-N were free to interact with these monosaccharides independently. CVN2 recognized ManNAc at a Kd of 1.4 μM and bound this sugar in solution, regardless of the lectin making amino acid side chain contacts on the targeted viral glycoproteins. An interdomain cross-contacting residue Glu41, which has been shown to be hydrogen bonding with dimannose, was substituted in the monomeric CV-N. The amide derivative of glucose, GlcNAc, achieved similar high affinity to the new variant CVN-E41T as high-mannose N-glycans, but binding to CVN2 in the nanomolar range with four binding sites involved or binding to the monomeric CVN-E41A. A stable dimer was engineered and expressed from the alanine-to-threonine-substituted monomer to confirm binding to GlcNAc. In summary, low-affinity binding was achieved by CVN2 to dimannosylated peptide or GlcNAc with two carbohydrate-binding sites of differing affinities, mimicking biological interactions with the respective N-linked glycans of interest and cross-linking of carbohydrates on human T cells for lymphocyte activation.
Collapse
Affiliation(s)
- Irene Maier
- Department
of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles, 650 Charles E. Young Dr. South, Los Angeles, California 90095, United States
- Department
of Internal Medicine I, Medical University
of Vienna, Waehringer Guertel 18-20, Vienna A-1090, Austria
| | - Georg Kontaxis
- Department
of Computational and Structural Biology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, Vienna 1030, Austria
| | - Christian Zimmermann
- Institute
of Chemical, Environmental and Bioscience Engineering, TU Wien, Gumpendorfer Strasse 1a, Wien 1060, Austria
| | - Christoph Steininger
- Department
of Internal Medicine I, Medical University
of Vienna, Waehringer Guertel 18-20, Vienna A-1090, Austria
| |
Collapse
|
3
|
Qiu Y, Wang H, Pan H, Ding X, Guan J, Zhuang Q, Wu K, Lei Z, Cai H, Dong Y, Zhou H, Lin A, Wang Q, Yan Q. NADH improves AIF dimerization and inhibits apoptosis in iPSCs-derived neurons from patients with auditory neuropathy spectrum disorder. Hear Res 2024; 441:108919. [PMID: 38043402 DOI: 10.1016/j.heares.2023.108919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 12/05/2023]
Abstract
Auditory neuropathy spectrum disorder (ANSD) is a hearing impairment involving disruptions to inner hair cells (IHCs), ribbon synapses, spiral ganglion neurons (SGNs), and/or the auditory nerve itself. The outcomes of cochlear implants (CI) for ANSD are variable and dependent on the location of lesion sites. Discovering a potential therapeutic agent for ANSD remains an urgent requirement. Here, 293T stable transfection cell lines and patient induced pluripotent stem cells (iPSCs)-derived auditory neurons carrying the apoptosis inducing factor (AIF) p.R422Q variant were used to pursue a therapeutic regent for ANSD. Nicotinamide adenine dinucleotide (NADH) is a main electron donor in the electron transport chain (ETC). In 293T stable transfection cells with the p.R422Q variant, NADH treatment improved AIF dimerization, rescued mitochondrial dysfunctions, and decreased cell apoptosis. The effects of NADH were further confirmed in patient iPSCs-derived neurons. The relative level of AIF dimers was increased to 150.7 % (P = 0.026) from 59.2 % in patient-neurons upon NADH treatment. Such increased AIF dimerization promoted the mitochondrial import of coiled-coil-helix-coiled-coil-helix domain-containing protein 4 (CHCHD4), which further restored mitochondrial functions. Similarly, the content of mitochondrial calcium (mCa2+) was downregulated from 136.7 % to 102.3 % (P = 0.0024) in patient-neurons upon NADH treatment. Such decreased mCa2+ levels inhibited calpain activity, ultimately reducing the percentage of apoptotic cells from 30.5 % to 21.1 % (P = 0.021). We also compared the therapeutic effects of gene correction and NADH treatment on hereditary ANSD. NADH treatment had comparable restorative effects on functions of ANSD patient-specific cells to that of gene correction. Our findings offer evidence of the molecular mechanisms of ANSD and introduce NADH as a potential therapeutic agent for ANSD therapy.
Collapse
Affiliation(s)
- Yue Qiu
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; Institute of Brain Science, Wannan Medical College, Wuhu, Anhui 241000, China
| | - Hongyang Wang
- Department of Audiology and Vestibular Medicine, Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, the Sixth Medicine Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Huaye Pan
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xue Ding
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jing Guan
- Department of Audiology and Vestibular Medicine, Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, the Sixth Medicine Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Qianqian Zhuang
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Kaiwen Wu
- Department of Audiology and Vestibular Medicine, Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, the Sixth Medicine Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Zhaoying Lei
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huajian Cai
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yufei Dong
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hui Zhou
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Aifu Lin
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qiuju Wang
- Department of Audiology and Vestibular Medicine, Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, the Sixth Medicine Center of Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | - Qingfeng Yan
- College of Life Science, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China; Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
4
|
Fazzari M, Di Biase E, Zaccagnini L, Henriques A, Callizot N, Ciampa MG, Mauri L, Carsana EV, Loberto N, Aureli M, Mari L, Civera M, Vasile F, Sonnino S, Bartels T, Chiricozzi E, Lunghi G. GM1 oligosaccharide efficacy against α-synuclein aggregation and toxicity in vitro. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159350. [PMID: 37330108 PMCID: PMC10579883 DOI: 10.1016/j.bbalip.2023.159350] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/11/2023] [Accepted: 06/01/2023] [Indexed: 06/19/2023]
Abstract
Fibrillary aggregated α-synuclein represents the neurologic hallmark of Parkinson's disease and is considered to play a causative role in the disease. Although the causes leading to α-synuclein aggregation are not clear, the GM1 ganglioside interaction is recognized to prevent this process. How GM1 exerts these functions is not completely clear, although a primary role of its soluble oligosaccharide (GM1-OS) is emerging. Indeed, we recently identified GM1-OS as the bioactive moiety responsible for GM1 neurotrophic and neuroprotective properties, specifically reverting the parkinsonian phenotype both in in vitro and in vivo models. Here, we report on GM1-OS efficacy against the α-synuclein aggregation and toxicity in vitro. By amyloid seeding aggregation assay and NMR spectroscopy, we demonstrated that GM1-OS was able to prevent both the spontaneous and the prion-like α-synuclein aggregation. Additionally, circular dichroism spectroscopy of recombinant monomeric α-synuclein showed that GM1-OS did not induce any change in α-synuclein secondary structure. Importantly, GM1-OS significantly increased neuronal survival and preserved neurite networks of dopaminergic neurons affected by α-synuclein oligomers, together with a reduction of microglia activation. These data further demonstrate that the ganglioside GM1 acts through its oligosaccharide also in preventing the α-synuclein pathogenic aggregation in Parkinson's disease, opening a perspective window for GM1-OS as drug candidate.
Collapse
Affiliation(s)
- Maria Fazzari
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy
| | - Erika Di Biase
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy
| | | | | | - Noëlle Callizot
- Neuro-Sys, 410 Chemin Départemental 60, 13120 Gardanne, France
| | - Maria Grazia Ciampa
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy
| | - Emma Veronica Carsana
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy
| | - Nicoletta Loberto
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy
| | - Luigi Mari
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Monica Civera
- Department of Chemistry, University of Milano, Milan, Italy
| | | | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy.
| | - Tim Bartels
- UK Dementia Research Institute at UCL, London, UK
| | - Elena Chiricozzi
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy.
| | - Giulia Lunghi
- Department of Medical Biotechnology and Translational Medicine, University of Milano, 20054 Segrate, Milano, Italy
| |
Collapse
|
5
|
Quintana JI, Atxabal U, Unione L, Ardá A, Jiménez-Barbero J. Exploring multivalent carbohydrate-protein interactions by NMR. Chem Soc Rev 2023; 52:1591-1613. [PMID: 36753338 PMCID: PMC9987413 DOI: 10.1039/d2cs00983h] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Indexed: 02/09/2023]
Abstract
Nuclear Magnetic Resonance (NMR) has been widely employed to assess diverse features of glycan-protein molecular recognition events. Different types of qualitative and quantitative information at different degrees of resolution and complexity can be extracted from the proper application of the available NMR-techniques. In fact, affinity, structural, kinetic, conformational, and dynamic characteristics of the binding process are available. Nevertheless, except in particular cases, the affinity of lectin-sugar interactions is weak, mostly at the low mM range. This feature is overcome in biological processes by using multivalency, thus augmenting the strength of the binding. However, the application of NMR methods to monitor multivalent lectin-glycan interactions is intrinsically challenging. It is well known that when large macromolecular complexes are formed, the NMR signals disappear from the NMR spectrum, due to the existence of fast transverse relaxation, related to the large size and exchange features. Indeed, at the heart of the molecular recognition event, the associated free-bound chemical exchange process for both partners takes place in a particular timescale. Thus, these factors have to be considered and overcome. In this review article, we have distinguished, in a subjective manner, the existence of multivalent presentations in the glycan or in the lectin. From the glycan perspective, we have also considered whether multiple epitopes of a given ligand are presented in the same linear chain of a saccharide (i.e., poly-LacNAc oligosaccharides) or decorating different arms of a multiantennae scaffold, either natural (as in multiantennae N-glycans) or synthetic (of dendrimer or polymer nature). From the lectin perspective, the presence of an individual binding site at every monomer of a multimeric lectin may also have key consequences for the binding event at different levels of complexity.
Collapse
Affiliation(s)
- Jon I Quintana
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain.
| | - Unai Atxabal
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain.
| | - Luca Unione
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain.
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Bizkaia, Spain
| | - Ana Ardá
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain.
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Bizkaia, Spain
| | - Jesús Jiménez-Barbero
- CICbioGUNE, Basque Research & Technology Alliance (BRTA), Bizkaia Technology Park, Building 800, 48160 Derio, Bizkaia, Spain.
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi 5, 48009 Bilbao, Bizkaia, Spain
- Department of Organic Chemistry, II Faculty of Science and Technology, EHU-UPV, 48940 Leioa, Spain
- Centro de Investigación Biomédica En Red de Enfermedades Respiratorias, Madrid, Spain
| |
Collapse
|
6
|
Caracciolo D, Juli G, Riillo C, Coricello A, Vasile F, Pollastri S, Rocca R, Scionti F, Polerà N, Grillone K, Arbitrio M, Staropoli N, Caparello B, Britti D, Loprete G, Costa G, Di Martino MT, Alcaro S, Tagliaferri P, Tassone P. Exploiting DNA Ligase III addiction of multiple myeloma by flavonoid Rhamnetin. Lab Invest 2022; 20:482. [PMID: 36273153 PMCID: PMC9588242 DOI: 10.1186/s12967-022-03705-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/11/2022] [Indexed: 11/28/2022]
Abstract
Background DNA ligases are crucial for DNA repair and cell replication since they catalyze the final steps in which DNA breaks are joined. DNA Ligase III (LIG3) exerts a pivotal role in Alternative-Non-Homologous End Joining Repair (Alt-NHEJ), an error-prone DNA repair pathway often up-regulated in genomically unstable cancer, such as Multiple Myeloma (MM). Based on the three-dimensional (3D) LIG3 structure, we performed a computational screening to identify LIG3-targeting natural compounds as potential candidates to counteract Alt-NHEJ activity in MM. Methods Virtual screening was conducted by interrogating the Phenol Explorer database. Validation of binding to LIG3 recombinant protein was performed by Saturation Transfer Difference (STD)—nuclear magnetic resonance (NMR) experiments. Cell viability was analyzed by Cell Titer-Glo assay; apoptosis was evaluated by flow cytometric analysis following Annexin V-7AAD staining. Alt-NHEJ repair modulation was evaluated using plasmid re-joining assay and Cytoscan HD. DNA Damage Response protein levels were analyzed by Western blot of whole and fractionated protein extracts and immunofluorescence analysis. The mitochondrial DNA (mtDNA) copy number was determined by qPCR. In vivo activity was evaluated in NOD-SCID mice subcutaneously engrafted with MM cells. Results Here, we provide evidence that a natural flavonoid Rhamnetin (RHM), selected by a computational approach, counteracts LIG3 activity and killed Alt-NHEJ-dependent MM cells. Indeed, Nuclear Magnetic Resonance (NMR) showed binding of RHM to LIG3 protein and functional experiments revealed that RHM interferes with LIG3-driven nuclear and mitochondrial DNA repair, leading to significant anti-MM activity in vitro and in vivo. Conclusion Taken together, our findings provide proof of concept that RHM targets LIG3 addiction in MM and may represent therefore a novel promising anti-tumor natural agent to be investigated in an early clinical setting. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03705-z.
Collapse
Affiliation(s)
- Daniele Caracciolo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Giada Juli
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Caterina Riillo
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Adriana Coricello
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | | | - Sara Pollastri
- Department of Chemistry, University of Milan, Milan, Italy
| | - Roberta Rocca
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Francesca Scionti
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), Messina, Italy
| | - Nicoletta Polerà
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Katia Grillone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Mariamena Arbitrio
- Institute of Research and Biomedical Innovation (IRIB), Italian National Council (CNR), Catanzaro, Italy
| | | | - Basilio Caparello
- Presidio Ospedaliero Giovanni Paolo II Lamezia Terme, Catanzaro, Italy
| | - Domenico Britti
- Department of Health Science, Magna Græcia University, Catanzaro, Italy
| | - Giovanni Loprete
- Department of Health Science, Magna Græcia University, Catanzaro, Italy
| | - Giosuè Costa
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Stefano Alcaro
- Department of Health Science, Magna Græcia University, Catanzaro, Italy.,Net4Science Academic Spin-Off, Magna Græcia University, Campus "Salvatore Venuta", Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy. .,Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Theillet FX, Luchinat E. In-cell NMR: Why and how? PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2022; 132-133:1-112. [PMID: 36496255 DOI: 10.1016/j.pnmrs.2022.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/19/2022] [Accepted: 04/27/2022] [Indexed: 06/17/2023]
Abstract
NMR spectroscopy has been applied to cells and tissues analysis since its beginnings, as early as 1950. We have attempted to gather here in a didactic fashion the broad diversity of data and ideas that emerged from NMR investigations on living cells. Covering a large proportion of the periodic table, NMR spectroscopy permits scrutiny of a great variety of atomic nuclei in all living organisms non-invasively. It has thus provided quantitative information on cellular atoms and their chemical environment, dynamics, or interactions. We will show that NMR studies have generated valuable knowledge on a vast array of cellular molecules and events, from water, salts, metabolites, cell walls, proteins, nucleic acids, drugs and drug targets, to pH, redox equilibria and chemical reactions. The characterization of such a multitude of objects at the atomic scale has thus shaped our mental representation of cellular life at multiple levels, together with major techniques like mass-spectrometry or microscopies. NMR studies on cells has accompanied the developments of MRI and metabolomics, and various subfields have flourished, coined with appealing names: fluxomics, foodomics, MRI and MRS (i.e. imaging and localized spectroscopy of living tissues, respectively), whole-cell NMR, on-cell ligand-based NMR, systems NMR, cellular structural biology, in-cell NMR… All these have not grown separately, but rather by reinforcing each other like a braided trunk. Hence, we try here to provide an analytical account of a large ensemble of intricately linked approaches, whose integration has been and will be key to their success. We present extensive overviews, firstly on the various types of information provided by NMR in a cellular environment (the "why", oriented towards a broad readership), and secondly on the employed NMR techniques and setups (the "how", where we discuss the past, current and future methods). Each subsection is constructed as a historical anthology, showing how the intrinsic properties of NMR spectroscopy and its developments structured the accessible knowledge on cellular phenomena. Using this systematic approach, we sought i) to make this review accessible to the broadest audience and ii) to highlight some early techniques that may find renewed interest. Finally, we present a brief discussion on what may be potential and desirable developments in the context of integrative studies in biology.
Collapse
Affiliation(s)
- Francois-Xavier Theillet
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France.
| | - Enrico Luchinat
- Dipartimento di Scienze e Tecnologie Agro-Alimentari, Alma Mater Studiorum - Università di Bologna, Piazza Goidanich 60, 47521 Cesena, Italy; CERM - Magnetic Resonance Center, and Neurofarba Department, Università degli Studi di Firenze, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
8
|
Abstract
In-cell structural biology aims at extracting structural information about proteins or nucleic acids in their native, cellular environment. This emerging field holds great promise and is already providing new facts and outlooks of interest at both fundamental and applied levels. NMR spectroscopy has important contributions on this stage: It brings information on a broad variety of nuclei at the atomic scale, which ensures its great versatility and uniqueness. Here, we detail the methods, the fundamental knowledge, and the applications in biomedical engineering related to in-cell structural biology by NMR. We finally propose a brief overview of the main other techniques in the field (EPR, smFRET, cryo-ET, etc.) to draw some advisable developments for in-cell NMR. In the era of large-scale screenings and deep learning, both accurate and qualitative experimental evidence are as essential as ever to understand the interior life of cells. In-cell structural biology by NMR spectroscopy can generate such a knowledge, and it does so at the atomic scale. This review is meant to deliver comprehensive but accessible information, with advanced technical details and reflections on the methods, the nature of the results, and the future of the field.
Collapse
Affiliation(s)
- Francois-Xavier Theillet
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198 Gif-sur-Yvette, France
| |
Collapse
|
9
|
Soares CO, Grosso AS, Ereño-Orbea J, Coelho H, Marcelo F. Molecular Recognition Insights of Sialic Acid Glycans by Distinct Receptors Unveiled by NMR and Molecular Modeling. Front Mol Biosci 2021; 8:727847. [PMID: 34869580 PMCID: PMC8634706 DOI: 10.3389/fmolb.2021.727847] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/01/2021] [Indexed: 11/13/2022] Open
Abstract
All cells are decorated with a highly dense and complex structure of glycan chains, which are mostly attached to proteins and lipids. In this context, sialic acids are a family of nine-carbon acidic monosaccharides typically found at the terminal position of glycan chains, modulating several physiological and pathological processes. Sialic acids have many structural and modulatory roles due to their negative charge and hydrophilicity. In addition, the recognition of sialic acid glycans by mammalian cell lectins, such as siglecs, has been described as an important immunological checkpoint. Furthermore, sialic acid glycans also play a pivotal role in host-pathogen interactions. Various pathogen receptors exposed on the surface of viruses and bacteria are responsible for the binding to sialic acid sugars located on the surface of host cells, becoming a critical point of contact in the infection process. Understanding the molecular mechanism of sialic acid glycans recognition by sialic acid-binding proteins, present on the surface of pathogens or human cells, is essential to realize the biological mechanism of these events and paves the way for the rational development of strategies to modulate sialic acid-protein interactions in diseases. In this perspective, nuclear magnetic resonance (NMR) spectroscopy, assisted with molecular modeling protocols, is a versatile and powerful technique to investigate the structural and dynamic aspects of glycoconjugates and their interactions in solution at the atomic level. NMR provides the corresponding ligand and protein epitopes, essential for designing and developing potential glycan-based therapies. In this review, we critically discuss the current state of knowledge about the structural features behind the molecular recognition of sialic acid glycans by different receptors, naturally present on human cells or pathogens, disclosed by NMR spectroscopy and molecular modeling protocols.
Collapse
Affiliation(s)
- Cátia Oliveira Soares
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal.,Department of Chemistry, UCIBIO-Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Ana Sofia Grosso
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal.,Department of Chemistry, UCIBIO-Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - June Ereño-Orbea
- CIC bioGUNE, Basque Research and Technology Alliance, Bizkaia Technology Park, Bilbao, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Helena Coelho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal.,Department of Chemistry, UCIBIO-Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| | - Filipa Marcelo
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal.,Department of Chemistry, UCIBIO-Applied Molecular Biosciences Unit, NOVA School of Science and Technology, NOVA University Lisbon, Caparica, Portugal
| |
Collapse
|
10
|
Della Volpe S, Linciano P, Listro R, Tumminelli E, Amadio M, Bonomo I, Elgaher WAM, Adam S, Hirsch AKH, Boeckler FM, Vasile F, Rossi D, Collina S. Identification of N,N-arylalkyl-picolinamide derivatives targeting the RNA-binding protein HuR, by combining biophysical fragment-screening and molecular hybridization. Bioorg Chem 2021; 116:105305. [PMID: 34482166 DOI: 10.1016/j.bioorg.2021.105305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/26/2021] [Accepted: 08/23/2021] [Indexed: 12/31/2022]
Abstract
Hu proteins are members of the RNA-binding protein (RBP) family and play a pivotal role in the regulation of post-transcriptional processes. Through interaction with selected mRNAs, RBPs regulate their function and stability; as a consequence, RBP dysregulation can cause abnormal translation of key proteins involved in several pathologies. In the past few years, this observation has sparked interest to develop new treatments against these pathologies by using small molecules able to modulate RBP activity. Among the four Hu proteins, we have directed our efforts towards the isoform HuR, which is mainly involved in cancer, inflammation and retinopathy. Aimed at developing compounds able to modulate the stability of HuR-mRNA complexes, in the present work, we applied a biophysical fragment screening by assessing a library of halogen-enriched heterocyclic fragments (HEFLibs) via Surface Plasmon Resonance (SPR) and Saturation Transfer Difference (STD) NMR to select promising fragments able to interact with HuR. One selected fragment and a few commercially available congeners were exploited to design and synthesize focused analogues of compound N-(3-chlorobenzyl)-N-(3,5-dihydroxyphenethyl)-4-hydroxybenzamide (1), our previously reported hit. STDNMR spectroscopy, molecular modeling, and SPR offered further insight into the HuR-small molecule interaction and showed that fragment-based approaches represent a promising and yet underexplored strategy to tackle such unusual targets. Lastly, fluorescence polarization (FP) studies revealed the capability of the new compounds to interfere with the formation of the HuR-mRNA complex. This is, to our knowledge, the first fragment-based campaign performed on the Hu protein class, and one of the few examples in the larger RBP field and constitutes an important step in the quest for the rational modulation of RBPs and related RNA functions by small molecules.
Collapse
Affiliation(s)
- S Della Volpe
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus building E8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany.
| | - P Linciano
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy.
| | - R Listro
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy.
| | - E Tumminelli
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy; Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus building E8.1, 66123 Saarbrücken, Germany.
| | - M Amadio
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy.
| | - I Bonomo
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.
| | - W A M Elgaher
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus building E8.1, 66123 Saarbrücken, Germany.
| | - S Adam
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus building E8.1, 66123 Saarbrücken, Germany.
| | - A K H Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Campus building E8.1, 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany.
| | - F M Boeckler
- Department of Pharmacy and Biochemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, Tübingen, Germany; Center for Bioinformatics Tübingen (ZBIT), Eberhard Karls Universität Tübingen, Tübingen, Germany.
| | - F Vasile
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy.
| | - D Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy.
| | - S Collina
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100, Pavia, Italy.
| |
Collapse
|
11
|
Lal K, Bermeo R, Cramer J, Vasile F, Ernst B, Imberty A, Bernardi A, Varrot A, Belvisi L. Prediction and Validation of a Druggable Site on Virulence Factor of Drug Resistant Burkholderia cenocepacia*. Chemistry 2021; 27:10341-10348. [PMID: 33769626 PMCID: PMC8360069 DOI: 10.1002/chem.202100252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Indexed: 12/12/2022]
Abstract
Burkholderia cenocepacia is an opportunistic Gram‐negative bacterium that causes infections in patients suffering from chronic granulomatous diseases and cystic fibrosis. It displays significant morbidity and mortality due to extreme resistance to almost all clinically useful antibiotics. The bacterial lectin BC2L‐C expressed in B. cenocepacia is an interesting drug target involved in bacterial adhesion and subsequent deadly infection to the host. We solved the first high resolution crystal structure of the apo form of the lectin N‐terminal domain (BC2L‐C‐nt) and compared it with the ones complexed with carbohydrate ligands. Virtual screening of a small fragment library identified potential hits predicted to bind in the vicinity of the fucose binding site. A series of biophysical techniques and X‐ray crystallographic screening were employed to validate the interaction of the hits with the protein domain. The X‐ray structure of BC2L‐C‐nt complexed with one of the identified active fragments confirmed the ability of the site computationally identified to host drug‐like fragments. The fragment affinity could be determined by titration microcalorimetry. These structure‐based strategies further provide an opportunity to elaborate the fragments into high affinity anti‐adhesive glycomimetics, as therapeutic agents against B. cenocepacia.
Collapse
Affiliation(s)
- Kanhaya Lal
- Universita' degli Studi di Milano, Dipartimento di Chimica, via Golgi 19, I-20133, Milano, Italy.,Université Grenoble Alpes, CNRS, CERMAV, 38000, Grenoble, France
| | - Rafael Bermeo
- Universita' degli Studi di Milano, Dipartimento di Chimica, via Golgi 19, I-20133, Milano, Italy.,Université Grenoble Alpes, CNRS, CERMAV, 38000, Grenoble, France
| | - Jonathan Cramer
- University of Basel, Department of Pharmaceutical Sciences, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Francesca Vasile
- Universita' degli Studi di Milano, Dipartimento di Chimica, via Golgi 19, I-20133, Milano, Italy
| | - Beat Ernst
- University of Basel, Department of Pharmaceutical Sciences, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Anne Imberty
- Université Grenoble Alpes, CNRS, CERMAV, 38000, Grenoble, France
| | - Anna Bernardi
- Universita' degli Studi di Milano, Dipartimento di Chimica, via Golgi 19, I-20133, Milano, Italy
| | - Annabelle Varrot
- Université Grenoble Alpes, CNRS, CERMAV, 38000, Grenoble, France
| | - Laura Belvisi
- Universita' degli Studi di Milano, Dipartimento di Chimica, via Golgi 19, I-20133, Milano, Italy
| |
Collapse
|
12
|
Ambrosio FA, Coricello A, Costa G, Lupia A, Micaelli M, Marchesi N, Sala F, Pascale A, Rossi D, Vasile F, Alcaro S, Collina S. Identification of Compounds Targeting HuD. Another Brick in the Wall of Neurodegenerative Disease Treatment. J Med Chem 2021; 64:9989-10000. [PMID: 34219450 DOI: 10.1021/acs.jmedchem.1c00191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
ELAV-like (ELAVL) RNA-binding proteins play a pivotal role in post-transcriptional processes, and their dysregulation is involved in several pathologies. This work was focused on HuD (ELAVL4), which is specifically expressed in nervous tissues, and involved in differentiation and synaptic plasticity mechanisms. HuD represents a new, albeit unexplored, candidate target for the treatment of several relevant neurodegenerative diseases. The aim of this pioneering work was the identification of new molecules able to recognize and bind HuD, thus interfering with its activity. We combined virtual screening, molecular dynamics (MD), and STD-NMR techniques. Starting from around 51 000 compounds, four promising hits eventually provided experimental evidence of their ability to bind HuD. Among the selected best hits, folic acid was found to be the most interesting one, being able to well recognize the HuD binding site. Our results provide a basis for the identification of new HuD interfering compounds which may be useful against neurodegenerative syndromes.
Collapse
Affiliation(s)
- Francesca Alessandra Ambrosio
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy
| | - Adriana Coricello
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy
| | - Giosuè Costa
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l'Innovazione Rurale, Località Condoleo, Belcastro, Catanzaro, Italy
| | - Antonio Lupia
- Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l'Innovazione Rurale, Località Condoleo, Belcastro, Catanzaro, Italy
| | - Mariachiara Micaelli
- CIBIO-Department of Cellular, Computational and Integrative Biology, University of Trento, Via Sommarive 9, Povo, 38123 Trento, Italy
| | - Nicoletta Marchesi
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Federico Sala
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy.,Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Alessia Pascale
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Francesca Vasile
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Campus "S. Venuta", 88100 Catanzaro, Italy.,Associazione CRISEA-Centro di Ricerca e Servizi Avanzati per l'Innovazione Rurale, Località Condoleo, Belcastro, Catanzaro, Italy
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
13
|
Phạm TTT, Rainey JK. On-cell nuclear magnetic resonance spectroscopy to probe cell surface interactions. Biochem Cell Biol 2021; 99:683-692. [PMID: 33945753 DOI: 10.1139/bcb-2021-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nuclear magnetic resonance (NMR) spectroscopy allows determination of atomic-level information about intermolecular interactions, molecular structure, and molecular dynamics in the cellular environment. This may be broadly divided into studies focused on obtaining detailed molecular information in the intracellular context ("in-cell") or those focused on characterizing molecules or events at the cell surface ("on-cell"). In this review, we outline some key NMR techniques applied for on-cell NMR studies through both solution-state and solid-state NMR and survey studies that have used these techniques to uncover key information. We particularly focus on application of on-cell NMR spectroscopy to characterize ligand interactions with cell surface membrane proteins such as G-protein coupled receptors (GPCRs), receptor tyrosine kinases, etc. These techniques allow for quantification of binding affinities, competitive binding assays, delineation of portions of ligands involved in binding, ligand bound-state conformational determination, evaluation of receptor structuring and dynamics, and inference of distance constraints characteristic of the ligand-receptor bound state. Excitingly, it is possible to avoid the barriers of production and purification of membrane proteins while obtaining directly physiologically-relevant information through on-cell NMR. We also provide a briefer survey of the applicability of on-cell NMR approaches to other classes of cell surface molecule.
Collapse
Affiliation(s)
- Trần Thanh Tâm Phạm
- Dalhousie University, 3688, Department of Biochemistry & Molecular Biology, Halifax, Nova Scotia, Canada;
| | - Jan K Rainey
- Dalhousie University, 3688, Department of Biochemistry & Molecular Biology, Halifax, Canada;
| |
Collapse
|
14
|
Wu K, Saha R, Su D, Krishna VD, Liu J, Cheeran MCJ, Wang JP. Magnetic-Nanosensor-Based Virus and Pathogen Detection Strategies before and during COVID-19. ACS APPLIED NANO MATERIALS 2020; 3:9560-9580. [PMID: 37556271 PMCID: PMC7526334 DOI: 10.1021/acsanm.0c02048] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/22/2020] [Indexed: 05/02/2023]
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes coronavirus disease 2019 (COVID-19), is a threat to the global healthcare system and economic security. As of July 2020, no specific drugs or vaccines are yet available for COVID-19; a fast and accurate diagnosis for SARS-CoV-2 is essential in slowing the spread of COVID-19 and for efficient implementation of control and containment strategies. Magnetic nanosensing is an emerging topic representing the frontiers of current biosensing and magnetic areas. The past decade has seen rapid growth in applying magnetic tools for biological and biomedical applications. Recent advances in magnetic nanomaterials and nanotechnologies have transformed current diagnostic methods to nanoscale and pushed the detection limit to early-stage disease diagnosis. Herein, this review covers the literature of magnetic nanosensors for virus and pathogen detection before COVID-19. We review popular magnetic nanosensing techniques including magnetoresistance, magnetic particle spectroscopy, and nuclear magnetic resonance. Magnetic point-of-care diagnostic kits are also reviewed aiming at developing plug-and-play diagnostics to manage the SARS-CoV-2 outbreak as well as preventing future epidemics. In addition, other platforms that use magnetic nanomaterials as auxiliary tools for enhanced pathogen and virus detection are also covered. The goal of this review is to inform the researchers of diagnostic and surveillance platforms for SARS-CoV-2 and their performances.
Collapse
Affiliation(s)
- Kai Wu
- Department of Electrical and Computer
Engineering, University of Minnesota,
Minneapolis, Minnesota 55455, United States
| | - Renata Saha
- Department of Electrical and Computer
Engineering, University of Minnesota,
Minneapolis, Minnesota 55455, United States
| | - Diqing Su
- Department of Chemical Engineering and
Material Science, University of Minnesota,
Minneapolis, Minnesota 55455, United States
| | - Venkatramana D. Krishna
- Department of Veterinary Population
Medicine, University of Minnesota, St.
Paul, Minnesota 55108, United States
| | - Jinming Liu
- Department of Electrical and Computer
Engineering, University of Minnesota,
Minneapolis, Minnesota 55455, United States
| | - Maxim C.-J. Cheeran
- Department of Veterinary Population
Medicine, University of Minnesota, St.
Paul, Minnesota 55108, United States
| | - Jian-Ping Wang
- Department of Electrical and Computer
Engineering, University of Minnesota,
Minneapolis, Minnesota 55455, United States
| |
Collapse
|
15
|
Civera M, Vasile F, Potenza D, Colombo C, Parente S, Vettraino C, Prosdocimi T, Parisini E, Belvisi L. Exploring E-cadherin-peptidomimetics interaction using NMR and computational studies. PLoS Comput Biol 2019; 15:e1007041. [PMID: 31158220 PMCID: PMC6564044 DOI: 10.1371/journal.pcbi.1007041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 06/13/2019] [Accepted: 04/22/2019] [Indexed: 02/02/2023] Open
Abstract
Cadherins are homophilic cell-cell adhesion molecules whose aberrant expression has often been shown to correlate with different stages of tumor progression. In this work, we investigate the interaction of two peptidomimetic ligands with the extracellular portion of human E-cadherin using a combination of NMR and computational techniques. Both ligands have been previously developed as mimics of the tetrapeptide sequence Asp1-Trp2-Val3-Ile4 of the cadherin adhesion arm, and have been shown to inhibit E-cadherin-mediated adhesion in epithelial ovarian cancer cells with millimolar potency. To sample a set of possible interactions of these ligands with the E-cadherin extracellular portion, STD-NMR experiments in the presence of two slightly different constructs, the wild type E-cadherin-EC1-EC2 fragment and the truncated E-cadherin-(Val3)-EC1-EC2 fragment, were carried out at three temperatures. Depending on the protein construct, a different binding epitope of the ligand and also a different temperature effect on STD signals were observed, both suggesting an involvement of the Asp1-Trp2 protein sequence among all the possible binding events. To interpret the experimental results at the atomic level and to probe the role of the cadherin adhesion arm in the dynamic interaction with the peptidomimetic ligand, a computational protocol based on docking calculations and molecular dynamics simulations was applied. In agreement with NMR data, the simulations at different temperatures unveil high variability/dynamism in ligand-cadherin binding, thus explaining the differences in ligand binding epitopes. In particular, the modulation of the signals seems to be dependent on the protein flexibility, especially at the level of the adhesive arm, which appears to participate in the interaction with the ligand. Overall, these results will help the design of novel cadherin inhibitors that might prevent the swap dimer formation by targeting both the Trp2 binding pocket and the adhesive arm residues. Classical cadherins are the main adhesive proteins at the intercellular junctions and play an essential role in tissue morphogenesis and homeostasis. A large number of studies have shown that cadherin aberrant expression and/or dysregulation often correlate with pathological processes, such as tumor development and progression. Notwithstanding the emerging role played by cadherins in a number of solid tumors, the rational design of small inhibitors targeting these proteins is still in its infancy, likely due to the challenges posed by the development of small drug-like molecules that modulate protein-protein interactions and to the structural complexity of the various cadherin dimerization interfaces that constantly form and disappear as the protein moves along its highly dynamic and reversible homo-dimerization trajectory. In this work, we study the interaction of two small molecules with the extracellular portion of human E-cadherin using a combination of spectroscopic and computational techniques. The availability of molecules interfering in the cadherin homophilic interactions could provide a useful tool for the investigation of cadherin function in tumors, and potentially pave the way to the development of novel alternative diagnostic and therapeutic interventions in cadherin-expressing solid tumors.
Collapse
Affiliation(s)
- Monica Civera
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
- Istituto di Scienze e Tecnologie Molecolari (ISTM), Consiglio Nazionale delle Ricerche, Milan, Italy
- * E-mail: (MC); (FV)
| | - Francesca Vasile
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
- Istituto di Scienze e Tecnologie Molecolari (ISTM), Consiglio Nazionale delle Ricerche, Milan, Italy
- * E-mail: (MC); (FV)
| | - Donatella Potenza
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | - Cinzia Colombo
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | - Sara Parente
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell'Insubria, Como, Italy
| | - Chiara Vettraino
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
| | - Tommaso Prosdocimi
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
| | - Emilio Parisini
- Center for Nano Science and Technology @PoliMi, Istituto Italiano di Tecnologia, Milan, Italy
| | - Laura Belvisi
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
- Istituto di Scienze e Tecnologie Molecolari (ISTM), Consiglio Nazionale delle Ricerche, Milan, Italy
| |
Collapse
|
16
|
Della Volpe S, Nasti R, Queirolo M, Unver MY, Jumde VK, Dömling A, Vasile F, Potenza D, Ambrosio FA, Costa G, Alcaro S, Zucal C, Provenzani A, Di Giacomo M, Rossi D, Hirsch AKH, Collina S. Novel Compounds Targeting the RNA-Binding Protein HuR. Structure-Based Design, Synthesis, and Interaction Studies. ACS Med Chem Lett 2019; 10:615-620. [PMID: 30996806 DOI: 10.1021/acsmedchemlett.8b00600] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/21/2019] [Indexed: 12/14/2022] Open
Abstract
The key role of RNA-binding proteins (RBPs) in regulating post-transcriptional processes and their involvement in several pathologies (i.e., cancer and neurodegeneration) have highlighted their potential as therapeutic targets. In this scenario, Embryonic Lethal Abnormal Vision (ELAV) or Hu proteins and their complexes with target mRNAs have been gaining growing attention. Compounds able to modulate the complex stability could constitute an innovative pharmacological strategy for the treatment of numerous diseases. Nevertheless, medicinal-chemistry efforts aimed at developing such compounds are still at an early stage. As part of our ongoing research in this field, we hereby present the rational design and synthesis of structurally novel HuR ligands, potentially acting as HuR-RNA interferers. The following assessment of the structural features of their interaction with HuR, combining saturation-transfer difference NMR and in silico studies, provides a guide for further research on the development of new effective interfering compounds of the HuR-RNA complex.
Collapse
Affiliation(s)
- Serena Della Volpe
- Department of Drug Sciences, Medicinal Chemistry and Technology Section, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Rita Nasti
- Department of Drug Sciences, Medicinal Chemistry and Technology Section, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Michele Queirolo
- Department of Drug Sciences, Medicinal Chemistry and Technology Section, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - M. Yagiz Unver
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization and Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, NL-9747 AG, Groningen, The Netherlands
| | - Varsha K. Jumde
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization and Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, NL-9747 AG, Groningen, The Netherlands
| | - Alexander Dömling
- Department of Drug Design, University of Groningen, A. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Francesca Vasile
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | - Donatella Potenza
- Department of Chemistry, University of Milan, Via Golgi 19, 20133 Milano, Italy
| | | | - Giosué Costa
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Stefano Alcaro
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Chiara Zucal
- Department of CIBIO, University of Trento, Via Sommarive 9, 38123 Povo, TN, Italy
| | | | - Marcello Di Giacomo
- Department of Drug Sciences, Medicinal Chemistry and Technology Section, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Daniela Rossi
- Department of Drug Sciences, Medicinal Chemistry and Technology Section, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| | - Anna K. H. Hirsch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization and Department of Pharmacy, Saarland University, Campus Building E8.1, 66123 Saarbrücken, Germany
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, NL-9747 AG, Groningen, The Netherlands
| | - Simona Collina
- Department of Drug Sciences, Medicinal Chemistry and Technology Section, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
17
|
Exploration of ligand binding modes towards the identification of compounds targeting HuR: a combined STD-NMR and Molecular Modelling approach. Sci Rep 2018; 8:13780. [PMID: 30214075 PMCID: PMC6137155 DOI: 10.1038/s41598-018-32084-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 08/29/2018] [Indexed: 12/18/2022] Open
Abstract
Post-transcriptional processes have been recognised as pivotal in the control of gene expression, and impairments in RNA processing are reported in several pathologies (i.e., cancer and neurodegeneration). Focusing on RNA-binding proteins (RBPs), the involvement of Embryonic Lethal Abnormal Vision (ELAV) or Hu proteins and their complexes with target mRNAs in the aetiology of various dysfunctions, has suggested the great potential of compounds able to interfere with the complex stability as an innovative pharmacological strategy for the treatment of numerous diseases. Here, we present a rational follow-up investigation of the interaction between ELAV isoform HuR and structurally-related compounds (i.e., flavonoids and coumarins), naturally decorated with different functional groups, by means of STD-NMR and Molecular Modelling. Our results represent the foundation for the development of potent and selective ligands able to interfere with ELAV–RNA complexes.
Collapse
|
18
|
Blaum BS, Neu U, Peters T, Stehle T. Spin ballet for sweet encounters: saturation-transfer difference NMR and X-ray crystallography complement each other in the elucidation of protein-glycan interactions. Acta Crystallogr F Struct Biol Commun 2018; 74:451-462. [PMID: 30084394 PMCID: PMC6096479 DOI: 10.1107/s2053230x18006581] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/28/2018] [Indexed: 03/11/2023] Open
Abstract
Biomolecular NMR spectroscopy has limitations in the determination of protein structures: an inherent size limit and the requirement for expensive and potentially difficult isotope labelling pose considerable hurdles. Therefore, structural analysis of larger proteins is almost exclusively performed by crystallography. However, the diversity of biological NMR applications outperforms that of any other structural biology technique. For the characterization of transient complexes formed by proteins and small ligands, notably oligosaccharides, one NMR technique has recently proven to be particularly powerful: saturation-transfer difference NMR (STD-NMR) spectroscopy. STD-NMR experiments are fast and simple to set up, with no general protein size limit and no requirement for isotope labelling. The method performs best in the moderate-to-low affinity range that is of interest in most of glycobiology. With small amounts of unlabelled protein, STD-NMR experiments can identify hits from mixtures of potential ligands, characterize mutant proteins and pinpoint binding epitopes on the ligand side. STD-NMR can thus be employed to complement and improve protein-ligand complex models obtained by other structural biology techniques or by purely computational means. With a set of protein-glycan interactions from our own work, this review provides an introduction to the technique for structural biologists. It exemplifies how crystallography and STD-NMR can be combined to elucidate protein-glycan (and other protein-ligand) interactions in atomic detail, and how the technique can extend structural biology from simplified systems amenable to crystallization to more complex biological entities such as membranes, live viruses or entire cells.
Collapse
Affiliation(s)
- Bärbel S. Blaum
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany
| | - Ursula Neu
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Thomas Peters
- Institute of Chemistry and Metabolomics, University of Lübeck, 23562 Lübeck, Germany
| | - Thilo Stehle
- Interfaculty Institute of Biochemistry, University of Tübingen, 72076 Tübingen, Germany
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| |
Collapse
|
19
|
Vasile F, Panigada M, Siccardi A, Potenza D, Tiana G. A Combined NMR-Computational Study of the Interaction between Influenza Virus Hemagglutinin and Sialic Derivatives from Human and Avian Receptors on the Surface of Transfected Cells. Int J Mol Sci 2018; 19:E1267. [PMID: 29695047 PMCID: PMC5983646 DOI: 10.3390/ijms19051267] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
The development of small-molecule inhibitors of influenza virus Hemagglutinin could be relevant to the opposition of the diffusion of new pandemic viruses. In this work, we made use of Nuclear Magnetic Resonance (NMR) spectroscopy to study the interaction between two derivatives of sialic acid, Neu5Ac-α-(2,6)-Gal-β-(1⁻4)-GlcNAc and Neu5Ac-α-(2,3)-Gal-β-(1⁻4)-GlcNAc, and hemagglutinin directly expressed on the surface of recombinant human cells. We analyzed the interaction of these trisaccharides with 293T cells transfected with the H5 and H1 variants of hemagglutinin, which thus retain their native trimeric conformation in such a realistic environment. By exploiting the magnetization transfer between the protein and the ligand, we obtained evidence of the binding event, and identified the epitope. We analyzed the conformational features of the glycans with an approach combining NMR spectroscopy and data-driven molecular dynamics simulations, thus obtaining useful information for an efficient drug design.
Collapse
Affiliation(s)
- Francesca Vasile
- Department of Chemistry, University of Milano, Via Golgi 19, 20133 Milano, Italy.
| | - Maddalena Panigada
- Molecular Immunology Unit, San Raffaele Research Institute, via Olgettina 58, 20132 Milano, Italy.
| | - Antonio Siccardi
- Molecular Immunology Unit, San Raffaele Research Institute, via Olgettina 58, 20132 Milano, Italy.
| | - Donatella Potenza
- Department of Chemistry, University of Milano, Via Golgi 19, 20133 Milano, Italy.
| | - Guido Tiana
- Center for Complexity and Biosystems and Department of Physics, University of Milano and INFN, Via Celoria 16, 20133 Milano, Italy.
| |
Collapse
|