1
|
Almeida GDO, Cintra ACO, Silva TA, de Oliveira IS, Correia LIV, Torquato RJS, Ferreira Junior RS, Arantes EC, Sampaio SV. Moojecin: The first disintegrin from Bothrops moojeni venom and its antitumor activity in acute myeloid leukemia. Int J Biol Macromol 2024; 279:135066. [PMID: 39197621 DOI: 10.1016/j.ijbiomac.2024.135066] [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: 05/14/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Disintegrins are a class of peptides found in snake venom that inhibit the activity of integrins, which are essential cell adhesion receptors in tumor progression and development. In this work, moojecin, a RGD disintegrin, was isolated from Bothrops moojeni snake venom, and its antitumor potential in acute myeloid leukemia (AML) HL-60 and THP-1 cells was characterized. The isolation was performed using a C18 reverse-phase column in two chromatographic steps, and its molecular mass is 7417.84 Da. N-terminal and de novo sequencing was performed to identify moojecin. Moojecin did not show cytotoxic or antiproliferative activity in THP-1 and HL-60 at tested concentrations, but it exhibited significant antimigratory activity in both cell lines, as well as inhibition of angiogenesis in the tube formation assay on Matrigel in a dose-dependent manner. A stronger interaction with integrin αVβ3 was shown in integrin interaction assays compared to α5β1, and the platelet aggregation assay indicated an IC50 of 5.039 μg/mL. Preliminary evaluation of disintegrin toxicity revealed no incidence of hemolysis or cytotoxic effects on peripheral blood mononuclear cells (PBMCs) across the tested concentrations. Thus, this is the first study to report the isolation, functional and structural characterization of a disintegrin from B. moojeni venom and bring a new perspective to assist in AML treatment.
Collapse
Affiliation(s)
- Gabriela de Oliveira Almeida
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Adélia Cristina Oliveira Cintra
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Thiago Abrahão Silva
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Isadora Sousa de Oliveira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | | | - Rui Seabra Ferreira Junior
- Center for the Study of Venoms and Venomous Animals (CEVAP), São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Eliane Candiani Arantes
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
2
|
Crack JC, Le Brun NE. Binding of a single nitric oxide molecule is sufficient to disrupt DNA binding of the nitrosative stress regulator NsrR. Chem Sci 2024:d4sc04618h. [PMID: 39464610 PMCID: PMC11500311 DOI: 10.1039/d4sc04618h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024] Open
Abstract
The regulatory protein NsrR, a member of the Rrf2 protein superfamily, plays a major role in the cellular response to nitrosative stress in many benign and pathogenic bacteria. The homodimeric protein binds a [4Fe-4S] cluster in each subunit (termed holo NsrR), and represses transcription of genes primarily involved in NO detoxification. Holo NsrR reacts rapidly with multiple NO molecules per [4Fe-4S] cluster, via a complex reaction, with loss of DNA binding and formation of NsrR-bound iron-nitrosyl species. However, the point at which DNA binding is lost is unknown. Here, we demonstrate using surface plasmon resonance (SPR) and native mass spectrometry (MS) that holo NsrR binds the promoter regions of NsrR-regulated genes with promoter-dependent nanomolar affinity, while hemi-apo NsrR (i.e. one cluster per dimer) binds >10-fold less tightly, and the cluster-free (apo) form not at all. Strikingly, native MS provided detailed information about the reaction of NO with the physiologically relevant form of NsrR, i.e. DNA-bound dimeric NsrR. Reaction with a single NO molecule per NsrR dimer is sufficient to abolish DNA binding. This exquisite sensitivity of DNA binding to NO is consistent with the importance of de-repressing NO detoxification systems at the earliest opportunity to minimise damage due to nitrosative stress. Furthermore, the data show that previously characterised iron-nitrosyls, which form at higher ratios of NO to [4Fe-4S], are not physiologically relevant for regulating the NsrR on/off switch.
Collapse
Affiliation(s)
- Jason C Crack
- Centre for Molecular and Structural Biochemistry, School of Chemistry, Pharmacy and Pharmacology, University of East Anglia Norwich Research Park Norwich NR4 7TJ UK
| | - Nick E Le Brun
- Centre for Molecular and Structural Biochemistry, School of Chemistry, Pharmacy and Pharmacology, University of East Anglia Norwich Research Park Norwich NR4 7TJ UK
| |
Collapse
|
3
|
Stocks BB, Thibeault MP, L'Abbé D, Umer M, Liu Y, Stuible M, Durocher Y, Melanson JE. Characterization of biotinylated human ACE2 and SARS-CoV-2 Omicron BA.4/5 spike protein reference materials. Anal Bioanal Chem 2024; 416:4861-4872. [PMID: 38942955 PMCID: PMC11330416 DOI: 10.1007/s00216-024-05413-7] [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: 02/29/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024]
Abstract
Accurate diagnostic and serology assays are required for the continued management of the COVID-19 pandemic yet spike protein mutations and intellectual property concerns with antigens and antibodies used in various test kits render comparability assessments difficult. As the use of common, well-characterized reagents can help address this lack of standardization, the National Research Council Canada has produced two protein reference materials (RMs) for use in SARS-CoV-2 serology assays: biotinylated human angiotensin-converting enzyme 2 RM, ACE2-1, and SARS-CoV-2 Omicron BA.4/5 spike protein RM, OMIC-1. Reference values were assigned through a combination of amino acid analysis via isotope dilution liquid chromatography tandem mass spectrometry following acid hydrolysis, and ultraviolet-visible (UV-Vis) spectrophotometry at 280 nm. Vial-to-vial homogeneity was established using UV-Vis measurements, and protein oligomeric status, monitored by size exclusion liquid chromatography (LC-SEC), was used to evaluate transportation, storage, and freeze-thaw stabilities. The molar protein concentration in ACE2-1 was 25.3 ± 1.7 µmol L-1 (k = 2, 95% CI) and consisted almost exclusively (98%) of monomeric ACE2, while OMIC-1 contained 5.4 ± 0.5 µmol L-1 (k = 2) spike protein in a mostly (82%) trimeric form. Glycoprotein molar mass determination by LC-SEC with multi-angle light scattering detection facilitated calculation of corresponding mass concentrations. To confirm protein functionality, the binding of OMIC-1 to immobilized ACE2-1 was investigated with surface plasmon resonance and the resulting dissociation constant, KD ~ 4.4 nM, was consistent with literature values.
Collapse
Affiliation(s)
- Bradley B Stocks
- Metrology, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada.
| | - Marie-Pier Thibeault
- Metrology, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Denis L'Abbé
- Human Health Therapeutics, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Muhammad Umer
- Metrology, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| | - Yali Liu
- Human Health Therapeutics, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Yves Durocher
- Human Health Therapeutics, National Research Council Canada, 6100 Royalmount Avenue, Montreal, QC, H4P 2R2, Canada
| | - Jeremy E Melanson
- Metrology, National Research Council Canada, 1200 Montreal Road, Ottawa, ON, K1A 0R6, Canada
| |
Collapse
|
4
|
Li K, Huntwork RH, Horn GQ, Alam SM, Tomaras GD, Dennison SM. TitrationAnalysis: a tool for high throughput binding kinetics data analysis for multiple label-free platforms. Gates Open Res 2024; 7:107. [PMID: 38009106 PMCID: PMC10667272 DOI: 10.12688/gatesopenres.14743.1] [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] [Accepted: 06/13/2024] [Indexed: 11/27/2023] Open
Abstract
Label-free techniques including Surface Plasmon Resonance (SPR) and Biolayer Interferometry (BLI) are biophysical tools widely used to collect binding kinetics data of bimolecular interactions. To efficiently analyze SPR and BLI binding kinetics data, we have built a new high throughput analysis tool named the TitrationAnalysis. It can be used as a package in the Mathematica scripting environment and ultilize the non-linear curve-fitting module of Mathematica for its core function. This tool can fit the binding time course data and estimate association and dissociation rate constants ( k a and k d respectively) for determining apparent dissociation constant ( K D ) values. The high throughput fitting process is automatic, requires minimal knowledge on Mathematica scripting and can be applied to data from multiple label-free platforms. We demonstrate that the TitrationAnalysis is optimal to analyze antibody-antigen binding data acquired on Biacore T200 (SPR), Carterra LSA (SPR imaging) and ForteBio Octet Red384 (BLI) platforms. The k a , k d and K D values derived using TitrationAnalysis very closely matched the results from the commercial analysis software provided specifically for these instruments. Additionally, the TitrationAnalysis tool generates user-directed customizable results output that can be readily used in downstream Data Quality Control associated with Good Clinical Laboratory Practice operations. With the versatility in source of data input source and options of analysis result output, the TitrationAnalysis high throughput analysis tool offers investigators a powerful alternative in biomolecular interaction characterization.
Collapse
Affiliation(s)
- Kan Li
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, 27701, USA
- Department of Surgery, Duke University, Durham, North Carolina, 27710, USA
| | - Richard H.C. Huntwork
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, 27701, USA
- Department of Surgery, Duke University, Durham, North Carolina, 27710, USA
| | - Gillian Q. Horn
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, 27701, USA
- Department of Surgery, Duke University, Durham, North Carolina, 27710, USA
| | - S. Munir Alam
- Department of Pathology, Duke University, Durham, North Carolina, 27710, USA
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, 27710, USA
| | - Georgia D. Tomaras
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, 27701, USA
- Department of Surgery, Duke University, Durham, North Carolina, 27710, USA
- Duke Human Vaccine Institute, Duke University, Durham, North Carolina, 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, 27710, USA
- Department of Integrative Immunobiology, Duke University, Durham, North Carolina, 27710, USA
| | - S. Moses Dennison
- Center for Human Systems Immunology, Duke University, Durham, North Carolina, 27701, USA
- Department of Surgery, Duke University, Durham, North Carolina, 27710, USA
| |
Collapse
|
5
|
Zhou M, Geng Z. Integrated LSPR Biosensing Signal Processing Strategy and Visualization Implementation. MICROMACHINES 2024; 15:631. [PMID: 38793204 PMCID: PMC11123047 DOI: 10.3390/mi15050631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024]
Abstract
The LSPR biosensor chip is a groundbreaking tool popular in laboratory settings for identifying disease markers. However, its use in clinical environments is not as widespread. One notable gap is the lack of a universal signal processing tool for LSPR biosensing. To escalate its precision, there is an emerging need for software that not only optimizes signal processing but also incorporates self-verification functionalities within LSPR biochemical sensors. Enter the visual LSPR sensor software-an innovative platform that processes real-time transmission or reflection spectra. This advanced software adeptly captures the nuanced structural changes at the nanostructure interface prompted by environmental fluctuations. It diligently records and computes a suite of parameters, including the resonance wavelength shift, full width at half maximum, sensitivity, and quality factor. These features empower users to tailor processing algorithms for each data capture session. Transcending traditional instruments, this method accommodates a multitude of parameters and ensures robust result validation while tactfully navigating nanostructure morphology complexities. Forsaking third-party tool dependencies, the software tackles challenges of precision and cost-effectiveness head-on, heralding a significant leap forward in nanophotonics, especially for high-throughput LSPR biosensing applications. This user-centric innovation marks substantial progress in biochemical detection. It is designed to serve both researchers and practitioners in the field of nanophotonic sensing technology, simplifying complexity while enhancing reliability and efficiency.
Collapse
Affiliation(s)
- Mixing Zhou
- School of Information Engineering, Minzu University of China, Beijing 100081, China;
| | - Zhaoxin Geng
- School of Information Engineering, Minzu University of China, Beijing 100081, China;
- Key Laboratory of Ethnic Language Intelligent Analysis and Security Governance of MOE, Minzu University of China, Beijing 100081, China
| |
Collapse
|
6
|
Wu HB, Wang CH, Chung YD, Shan YS, Lin YJ, Tsai HP, Lee GB. Highly-specific aptamer targeting SARS-CoV-2 S1 protein screened on an automatic integrated microfluidic system for COVID-19 diagnosis. Anal Chim Acta 2023; 1274:341531. [PMID: 37455073 DOI: 10.1016/j.aca.2023.341531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
Variants of the severe acute respiratory syndrome coronavirus (SARS-CoV-2) have evolved such that it may be challenging for diagnosis and clinical treatment of the pandemic coronavirus disease-19 (COVID-19). Compared with developed SARS-CoV-2 diagnostic tools recently, aptamers may exhibit some advantages, including high specificity/affinity, longer shelf life (vs. antibodies), and could be easily prepared. Herein an integrated microfluidic system was developed to automatically carry out one novel screening process based on the systematic evolution of ligands by exponential enrichment (SELEX) for screening aptamers specific with SARS-CoV-2. The new screening process started with five rounds of positive selection (with the S1 protein of SARS-CoV-2). In addition, including non-target viruses (influenza A and B), human respiratory tract-related cancer cells (adenocarcinoma human alveolar basal epithelial cells and dysplastic oral keratinocytes), and upper respiratory tract-related infectious bacteria (including methicillin-resistant Staphylococcus aureus, Pseudomonas aeruginosa, Acinetobacter baumannii, and Klebsiella pneumoniae), and human saliva were involved to increase the specificity of the screened aptamer during the negative selection. Totally, all 10 rounds could be completed within 20 h. The dissociation constant of the selected aptamer was determined to be 63.0 nM with S1 protein. Limits of detection for Wuhan and Omicron clinical strains were found to be satisfactory for clinical applications (i.e. 4.80 × 101 and 1.95 × 102 copies/mL, respectively). Moreover, the developed aptamer was verified to be capable of capturing inactivated SARS-CoV-2 viruses, eight SARS-CoV-2 pseudo-viruses, and clinical isolates of SARS-CoV-2 viruses. For high-variable emerging viruses, this developed integrated microfluidic system can be used to rapidly select highly-specific aptamers based on the novel SELEX methods to deal with infectious diseases in the future.
Collapse
Affiliation(s)
- Hung-Bin Wu
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Hung Wang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Da Chung
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan; Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ying-Jun Lin
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Huey-Pin Tsai
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Gwo-Bin Lee
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu, Taiwan; Institute of NanoEngineering and Microsystems, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
7
|
Nicastro G, Abis G, Klein P, Esteban-Serna S, Gallagher C, Chaves-Arquero B, Cai Y, Figueiredo AM, Martin SR, Patani R, Taylor IA, Ramos A. Direct m6A recognition by IMP1 underlays an alternative model of target selection for non-canonical methyl-readers. Nucleic Acids Res 2023; 51:8774-8786. [PMID: 37377445 PMCID: PMC10484666 DOI: 10.1093/nar/gkad534] [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: 02/24/2023] [Revised: 06/02/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
m6A methylation provides an essential layer of regulation in organismal development, and is aberrant in a range of cancers and neuro-pathologies. The information encoded by m6A methylation is integrated into existing RNA regulatory networks by RNA binding proteins that recognise methylated sites, the m6A readers. m6A readers include a well-characterised class of dedicated proteins, the YTH proteins, as well as a broader group of multi-functional regulators where recognition of m6A is only partially understood. Molecular insight in this recognition is essential to build a mechanistic understanding of global m6A regulation. In this study, we show that the reader IMP1 recognises the m6A using a dedicated hydrophobic platform that assembles on the methyl moiety, creating a stable high-affinity interaction. This recognition is conserved across evolution and independent from the underlying sequence context but is layered upon the strong sequence specificity of IMP1 for GGAC RNA. This leads us to propose a concept for m6A regulation where methylation plays a context-dependent role in the recognition of selected IMP1 targets that is dependent on the cellular concentration of available IMP1, differing from that observed for the YTH proteins.
Collapse
Affiliation(s)
- Giuseppe Nicastro
- Macromolecular Structure Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Giancarlo Abis
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, UK
| | - Pierre Klein
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, UK
| | - Sofia Esteban-Serna
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, UK
| | - Christopher Gallagher
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, UK
| | - Belen Chaves-Arquero
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, UK
| | - Yuyang Cai
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, UK
| | - Angelo Miguel Figueiredo
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, UK
| | - Stephen R Martin
- Structural Biology Technology Platform, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Rickie Patani
- Human Stem Cells and Neurodegeneration Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ian A Taylor
- Macromolecular Structure Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Andres Ramos
- Division of Biosciences, Institute of Structural and Molecular Biology, University College London, London, UK
| |
Collapse
|
8
|
Helsen C, Nguyen TT, Lee XY, Eerlings R, Louros N, Schymkowitz J, Rousseau F, Claessens F, Voet A. Exploiting Ligand-binding Domain Dimerization for Development of Novel Androgen Receptor Inhibitors. Mol Cancer Ther 2022; 21:1823-1834. [PMID: 36218067 DOI: 10.1158/1535-7163.mct-22-0340] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/07/2022] [Accepted: 10/03/2022] [Indexed: 01/12/2023]
Abstract
Currently, all clinically used androgen receptor (AR) antagonists target the AR ligand-binding pocket and inhibit T and dihydrotestosterone (DHT) binding. Resistance to these inhibitors in prostate cancer frequently involves AR-dependent mechanisms resulting in a retained AR dependence of the tumor. More effective or alternative AR inhibitors are therefore required to limit progression in these resistant stages. Here, we applied the structural information of the ligand-binding domain (LBD) dimerization interface to screen in silico for inhibitors. A completely new binding site, the Dimerisation Inhibiting Molecules (DIM) pocket, was identified at the LBD dimerization interface. Selection of compounds that fit the DIM pocket via virtual screening identified the DIM20 family of compounds which inhibit AR transactivation and dimerization of the full-length AR as well as the isolated LBDs. Via biolayer interferometry, reversible dose-dependent binding to the LBD was confirmed. While DIM20 does not compete with 3H-DHT for binding in the LBP, it limits the maximal activity of the AR indicative of a noncompetitive binding to the LBD. DIM20 and DIM20.39 specifically inhibit proliferation of AR-positive prostate cancer cell lines, with only marginal effects on AR-negative cell lines such as HEK 293 and PC3. Moreover, combination treatment of DIM compounds with enzalutamide results in synergistic antiproliferative effects which underline the specific mechanism of action of the DIM compounds.
Collapse
Affiliation(s)
- Christine Helsen
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Tien T Nguyen
- Laboratory of Biomolecular Modelling and Design, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Xiao Yin Lee
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Roy Eerlings
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nikolaos Louros
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Arnout Voet
- Laboratory of Biomolecular Modelling and Design, Department of Chemistry, KU Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Wang L, Wan Y, Ma N, Zhou L, Zhao D, Yu J, Wang H, Lin Z, Qian W. Real-time kinetics and affinity analysis of the interaction between protein A and immunoglobulins G derived from different species on silica colloidal crystal films. Colloids Surf B Biointerfaces 2022; 219:112839. [PMID: 36137338 DOI: 10.1016/j.colsurfb.2022.112839] [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: 07/19/2022] [Revised: 08/24/2022] [Accepted: 09/10/2022] [Indexed: 10/31/2022]
Abstract
Kinetic and affinity analysis of protein interactions reveals information on their related activities in biological processes. Herein, we established a system for evaluating the kinetics and affinity of the interaction between protein A and various IgG species on the surface of silica spheres of silica colloidal crystal (SCC) films by the extraordinary optical interference capabilities of 190 nm silica spheres after self-assembly. The equilibrium association constant (KA) was calculated by the equilibrium Langmuir model and nonlinear least-squares analysis of time-dependent data. The relative protein A/IgG binding affinity is human > rabbit >cow >goat. In addition, the competitive interaction of distinct species of IgG with protein A at the interface of SCC films was studied and performed. These findings may help with the use of protein A and other recognition components in a number of sensor types. Furthermore, this research might offer a novel approach to determining the kinetics and affinity of proteins on the surface of spheres particles, which may contribute to the development of the application of spheres particles in pharmaceutical science, biomedical engineering, and other techniques.
Collapse
Affiliation(s)
- Lu Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yizhen Wan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ning Ma
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lele Zhou
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Dongmin Zhao
- Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Jianning Yu
- Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Huili Wang
- Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Zhiping Lin
- Nanjing Weigang Dairy Co., Ltd., Nanjing 211102, China
| | - Weiping Qian
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| |
Collapse
|
10
|
Santos JC, Dametto M, Masson AP, Faça VM, Bonacin R, Donadi EA, Passos GA. The AIRE G228W mutation disturbs the interaction of AIRE with its partner molecule SIRT1. Front Immunol 2022; 13:948419. [PMID: 36148232 PMCID: PMC9485725 DOI: 10.3389/fimmu.2022.948419] [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: 05/19/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
The autoimmune regulator (AIRE) protein functions as a tetramer, interacting with partner proteins to form the “AIRE complex,” which relieves RNA Pol II stalling in the chromatin of medullary thymic epithelial cells (mTECs). AIRE is the primary mTEC transcriptional controller, promoting the expression of a large set of peripheral tissue antigen genes implicated in the negative selection of self-reactive thymocytes. Under normal conditions, the SIRT1 protein temporarily interacts with AIRE and deacetylates K residues of the AIRE SAND domain. Once the AIRE SAND domain is deacetylated, the binding with SIRT1 is undone, allowing the AIRE complex to proceed downstream with the RNA Pol II to the elongation phase of transcription. Considering that the in silico and in vitro binding of the AIRE SAND domain with SIRT1 provides a powerful model system for studying the dominant SAND G228W mutation mechanism, which causes the autoimmune polyglandular syndrome-1, we integrated computational molecular modeling, docking, dynamics between the whole SAND domain with SIRT1, and surface plasmon resonance using a peptide harboring the 211 to 230 residues of the SAND domain, to compare the structure and energetics of binding/release between AIRE G228 (wild-type) and W228 (mutant) SAND domain to SIRT1. We observed that the G228W mutation in the SAND domain negatively influences the AIRE-SIRT1 interaction. The disturbed interaction might cause a disruption in the binding of the AIRE SAND domain with the SIRT1 catalytic site, impairing the AIRE complex to proceed downstream with RNA Pol II.
Collapse
Affiliation(s)
- Jadson C. Santos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Mariangela Dametto
- Renato Archer Information Technology Center (CTI Brazil), Ministry of Science, Technology and Innovation (MCTI), Campinas, SP, Brazil
| | - Ana Paula Masson
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Vitor M. Faça
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Rodrigo Bonacin
- Renato Archer Information Technology Center (CTI Brazil), Ministry of Science, Technology and Innovation (MCTI), Campinas, SP, Brazil
| | - Eduardo A. Donadi
- Division of Clinical Immunology, Department of Medicine, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Geraldo Aleixo Passos
- Molecular Immunogenetics Group, Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- Laboratory of Genetics and Molecular Biology, Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- Center for Cell-Based Therapy in Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
- *Correspondence: Geraldo Aleixo Passos,
| |
Collapse
|
11
|
Moreira G, Casso-Hartmann L, Datta SPA, Dean D, McLamore E, Vanegas D. Development of a Biosensor Based on Angiotensin-Converting Enzyme II for Severe Acute Respiratory Syndrome Coronavirus 2 Detection in Human Saliva. FRONTIERS IN SENSORS 2022; 3:917380. [PMID: 35992634 PMCID: PMC9386735 DOI: 10.3389/fsens.2022.917380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the novel coronavirus responsible for COVID-19. Infection in humans requires angiotensin-converting enzyme II (hACE2) as the point of entry for SARS-CoV-2. PCR testing is generally definitive but expensive, although it is highly sensitive and accurate. Biosensor-based monitoring could be a low-cost, accurate, and non-invasive approach to improve testing capacity. We develop a capacitive hACE2 biosensor for intact SARS-CoV-2 detection in saliva. Laser-induced graphene (LIG) electrodes were modified with platinum nanoparticles. The quality control of LIG electrodes was performed using cyclic voltammetry. Truncated hACE2 was used as a biorecognition element and attached to the electrode surface by streptavidin-biotin coupling. Biolayer interferometry was used for qualitative interaction screening of hACE2 with UV-attenuated virions. Electrochemical impedance spectroscopy (EIS) was used for signal transduction. Truncated hACE2 binds wild-type SARS-CoV-2 and its variants with greater avidity than human coronavirus (common cold virus). The limit of detection (LoD) is estimated to be 2,960 copies/ml. The detection process usually takes less than 30 min. The strength of these features makes the hACE2 biosensor a potentially low-cost approach for screening SARS-CoV-2 in non-clinical settings with high demand for rapid testing (for example, schools and airports).
Collapse
Affiliation(s)
- Geisianny Moreira
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC, United States
- Global Alliance for Rapid Diagnostics, Michigan State University, Cambridge, MI, United States
| | - Lisseth Casso-Hartmann
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC, United States
| | - Shoumen Palit Austin Datta
- Medical Device (MDPnP) Interoperability and Cybersecurity Labs, Biomedical Engineering Program, Department of Anesthesiology, Massachusetts General Hospital, Harvard Medical School, Cambridge, MA, United States
- MIT Auto-ID Labs, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Delphine Dean
- Center for Innovative Medical Devices and Sensors (REDDI Lab), Clemson University, Clemson, SC, United States
- Department of Bioengineering, Clemson University, Clemson, SC, United States
| | - Eric McLamore
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC, United States
- Global Alliance for Rapid Diagnostics, Michigan State University, Cambridge, MI, United States
- Department of Agricultural Sciences, Clemson University, Clemson, SC, United States
| | - Diana Vanegas
- Department of Environmental Engineering and Earth Sciences, Clemson University, Clemson, SC, United States
- Global Alliance for Rapid Diagnostics, Michigan State University, Cambridge, MI, United States
| |
Collapse
|
12
|
Skokowa J, Hernandez Alvarez B, Coles M, Ritter M, Nasri M, Haaf J, Aghaallaei N, Xu Y, Mir P, Krahl AC, Rogers KW, Maksymenko K, Bajoghli B, Welte K, Lupas AN, Müller P, ElGamacy M. A topological refactoring design strategy yields highly stable granulopoietic proteins. Nat Commun 2022; 13:2948. [PMID: 35618709 PMCID: PMC9135769 DOI: 10.1038/s41467-022-30157-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 04/19/2022] [Indexed: 11/09/2022] Open
Abstract
Protein therapeutics frequently face major challenges, including complicated production, instability, poor solubility, and aggregation. De novo protein design can readily address these challenges. Here, we demonstrate the utility of a topological refactoring strategy to design novel granulopoietic proteins starting from the granulocyte-colony stimulating factor (G-CSF) structure. We change a protein fold by rearranging the sequence and optimising it towards the new fold. Testing four designs, we obtain two that possess nanomolar activity, the most active of which is highly thermostable and protease-resistant, and matches its designed structure to atomic accuracy. While the designs possess starkly different sequence and structure from the native G-CSF, they show specific activity in differentiating primary human haematopoietic stem cells into mature neutrophils. The designs also show significant and specific activity in vivo. Our topological refactoring approach is largely independent of sequence or structural context, and is therefore applicable to a wide range of protein targets. Skokowa et al. reconstruct the fold of a granulopoietic cytokine, resulting in de novo, hyperstable, highly active proteins with therapeutic potential for treating several neutropenia disorders.
Collapse
Affiliation(s)
- Julia Skokowa
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany.
| | | | - Murray Coles
- Max Planck Institute for Biology, 72076, Tübingen, Germany
| | - Malte Ritter
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Masoud Nasri
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Jérémy Haaf
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Narges Aghaallaei
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Yun Xu
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Perihan Mir
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Ann-Christin Krahl
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Katherine W Rogers
- Friedrich Miescher Laboratory of the Max Planck Society, 72076, Tübingen, Germany.,Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Kateryna Maksymenko
- Max Planck Institute for Biology, 72076, Tübingen, Germany.,Friedrich Miescher Laboratory of the Max Planck Society, 72076, Tübingen, Germany
| | - Baubak Bajoghli
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Karl Welte
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Andrei N Lupas
- Max Planck Institute for Biology, 72076, Tübingen, Germany
| | - Patrick Müller
- Friedrich Miescher Laboratory of the Max Planck Society, 72076, Tübingen, Germany.,Department of Biology, University of Konstanz, 78464, Konstanz, Germany
| | - Mohammad ElGamacy
- Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, 72076, Tübingen, Germany. .,Friedrich Miescher Laboratory of the Max Planck Society, 72076, Tübingen, Germany. .,Heliopolis Biotechnology Ltd, Cambridge, CB24 9RX, UK. .,Max Planck Institute for Biology, 72076, Tübingen, Germany.
| |
Collapse
|
13
|
Engel H, Guischard F, Krause F, Nandy J, Kaas P, Höfflin N, Köhn M, Kilb N, Voigt K, Wolf S, Aslan T, Baezner F, Hahne S, Ruckes C, Weygant J, Zinina A, Akmeriç EB, Antwi EB, Dombrovskij D, Franke P, Lesch KL, Vesper N, Weis D, Gensch N, Di Ventura B, Öztürk MA. finDr: A web server for in silico D-peptide ligand identification. Synth Syst Biotechnol 2021; 6:402-413. [PMID: 34901479 PMCID: PMC8632724 DOI: 10.1016/j.synbio.2021.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/20/2021] [Accepted: 11/08/2021] [Indexed: 11/18/2022] Open
Abstract
In the rapidly expanding field of peptide therapeutics, the short in vivo half-life of peptides represents a considerable limitation for drug action. D-peptides, consisting entirely of the dextrorotatory enantiomers of naturally occurring levorotatory amino acids (AAs), do not suffer from these shortcomings as they are intrinsically resistant to proteolytic degradation, resulting in a favourable pharmacokinetic profile. To experimentally identify D-peptide binders to interesting therapeutic targets, so-called mirror-image phage display is typically performed, whereby the target is synthesized in D-form and L-peptide binders are screened as in conventional phage display. This technique is extremely powerful, but it requires the synthesis of the target in D-form, which is challenging for large proteins. Here we present finDr, a novel web server for the computational identification and optimization of D-peptide ligands to any protein structure (https://findr.biologie.uni-freiburg.de/). finDr performs molecular docking to virtually screen a library of helical 12-mer peptides extracted from the RCSB Protein Data Bank (PDB) for their ability to bind to the target. In a separate, heuristic approach to search the chemical space of 12-mer peptides, finDr executes a customizable evolutionary algorithm (EA) for the de novo identification or optimization of D-peptide ligands. As a proof of principle, we demonstrate the validity of our approach to predict optimal binders to the pharmacologically relevant target phenol soluble modulin alpha 3 (PSMα3), a toxin of methicillin-resistant Staphylococcus aureus (MRSA). We validate the predictions using in vitro binding assays, supporting the success of this approach. Compared to conventional methods, finDr provides a low cost and easy-to-use alternative for the identification of D-peptide ligands against protein targets of choice without size limitation. We believe finDr will facilitate D-peptide discovery with implications in biotechnology and biomedicine.
Collapse
Key Words
- D-AA, dextrorotatory amino acid
- D-peptide
- EA, evolutionary algorithm
- Evolutionary algorithm
- L-AA, levorotatory amino acid
- MD, molecular dynamics
- MIEA, mirror-image evolutionary algorithm
- MIPD, mirror-image phage display
- MIVS, mirror-image virtual screening
- MRSA, methicillin-resistant Staphylococcus aureus
- Mirror-image phage display
- Molecular docking
- NCL, native chemical ligation
- PD-1, receptor programmed death 1
- PPI, protein-protein interaction
- PSMα3, phenol soluble modulin alpha 3
- Peptide design
- SPPS, solid phase peptide synthesis
- Web server
Collapse
Affiliation(s)
- Helena Engel
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Felix Guischard
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Fabian Krause
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Janina Nandy
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Paulina Kaas
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Nico Höfflin
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestr. 1, 79104, Freiburg, Germany
| | - Maja Köhn
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestr. 1, 79104, Freiburg, Germany
| | - Normann Kilb
- Institute of Biology II, Faculty of Biology, University of Freiburg, Schänzlestr. 1, 79104, Freiburg, Germany
- AG Roth-Lab for MicroarrayCopying, ZBSA–Centre for Biological Systems Analysis, University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany
| | - Karsten Voigt
- Institute of Biology III, Faculty of Biology, University of Freiburg, Schänzlestr. 1, 79104, Freiburg, Germany
| | - Steffen Wolf
- Biomolecular Dynamics, Institute of Physics, University of Freiburg, Hermann-Herder-Strasse 3a, 79104, Freiburg, Germany
| | - Tahira Aslan
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Fabian Baezner
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Salomé Hahne
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Carolin Ruckes
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Joshua Weygant
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Alisa Zinina
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Emir Bora Akmeriç
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Enoch B. Antwi
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Dennis Dombrovskij
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Philipp Franke
- Institute for Biochemistry, University of Freiburg, Albertstr. 21, 79104, Freiburg, Germany
| | - Klara L. Lesch
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
- Institute of Biology II, Faculty of Biology, University of Freiburg, Schänzlestr. 1, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19A, 79104, Freiburg, Germany
- Internal Medicine IV, Department of Medicine, Medical Center, University of Freiburg, Hugstetter Straße 55, 79106, Freiburg, Germany
| | - Niklas Vesper
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Daniel Weis
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Nicole Gensch
- Core Facility Signalling Factory, Centre for Biological Signaling Studies (BIOSS), University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
- Corresponding author. Core Facility Signalling Factory, Centre for Biological Signaling Studies (BIOSS), University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany.
| | - Barbara Di Ventura
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
- Institute of Biology II, Faculty of Biology, University of Freiburg, Schänzlestr. 1, 79104, Freiburg, Germany
- Corresponding author. Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany.
| | - Mehmet Ali Öztürk
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
- Institute of Biology II, Faculty of Biology, University of Freiburg, Schänzlestr. 1, 79104, Freiburg, Germany
- Corresponding author. Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany.
| |
Collapse
|
14
|
Björklund E, du Rietz A, Lundström P. Analysis of protein-ligand interactions from titrations and nuclear magnetic resonance relaxation dispersions. Protein Sci 2021; 31:301-307. [PMID: 34791737 PMCID: PMC8740844 DOI: 10.1002/pro.4240] [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: 07/31/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/22/2022]
Abstract
We present PLIS, a publicly available, open‐source software for the determination of protein–ligand dissociation constants that can be used to characterize biological processes or to shed light on biophysical aspects of interactions. PLIS can analyze data from titration experiments monitored by for instance fluorescence spectroscopy or from nuclear magnetic resonance relaxation dispersion experiments. In addition to analysis of experimental data, PLIS includes functionality for generation of synthetic data, useful for understanding how different parameters effect the data in order to better analyze experiments.
Collapse
Affiliation(s)
- Emil Björklund
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Anna du Rietz
- Division of Molecular Surface Physics and Nanoscience, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Patrik Lundström
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| |
Collapse
|
15
|
Chaves-Arquero B, Collins KM, Christodoulou E, Nicastro G, Martin SR, Ramos A. The distinct RNA-interaction modes of a small ZnF domain underlay TUT4(7) diverse action in miRNA regulation. RNA Biol 2021; 18:770-781. [PMID: 34719327 PMCID: PMC8782169 DOI: 10.1080/15476286.2021.1991169] [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] [Indexed: 10/24/2022] Open
Abstract
TUT4 and the closely related TUT7 are non-templated poly(U) polymerases required at different stages of development, and their mis-regulation or mutation has been linked to important cancer pathologies. While TUT4(7) interaction with its pre-miRNA targets has been characterized in detail, the molecular bases of the broader target recognition process are unclear. Here, we examine RNA binding by the ZnF domains of the protein. We show that TUT4(7) ZnF2 contains two distinct RNA binding surfaces that are used in the interaction with different RNA nucleobases in different targets, i.e that this small domain encodes diversity in TUT4(7) selectivity and molecular function. Interestingly and unlike other well-characterized CCHC ZnFs, ZnF2 is not physically coupled to the flanking ZnF3 and acts independently in miRNA recognition, while the remaining CCHC ZnF of TUT4(7), ZnF1, has lost its intrinsic RNA binding capability. Together, our data suggest that the ZnFs of TUT4(7) are independent units for RNA and, possibly, protein-protein interactions that underlay the protein's functional flexibility and are likely to play an important role in building its interaction network.
Collapse
Affiliation(s)
- Belén Chaves-Arquero
- Institute of Structural and Molecular Biology (ISMB) instead of (Ismb), University College London, London, UK
| | - Katherine M Collins
- Institute of Structural and Molecular Biology (ISMB) instead of (Ismb), University College London, London, UK
| | | | - Giuseppe Nicastro
- Macromolecular Structure Laboratory, The Francis Crick Institute, London, UK
| | - Stephen R Martin
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Andres Ramos
- Institute of Structural and Molecular Biology (ISMB) instead of (Ismb), University College London, London, UK
| |
Collapse
|
16
|
Conrad M, Fechner P, Proll G, Gauglitz G. Comparison of methods for quantitative biomolecular interaction analysis. Anal Bioanal Chem 2021; 414:661-673. [PMID: 34505164 PMCID: PMC8748344 DOI: 10.1007/s00216-021-03623-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 11/24/2022]
Abstract
In order to perform good kinetic experiments, not only the experimental conditions have to be optimized, but the evaluation procedure as well. The focus of this work is the in-depth comparison of different approaches and algorithms to determine kinetic rate constants for biomolecular interaction analysis (BIA). The different algorithms are applied not only to flawless simulated data, but also to real-world measurements. We compare five mathematical approaches for the evaluation of binding curves following pseudo-first-order kinetics with different noise levels. In addition, reflectometric interference spectroscopy (RIfS) measurements of two antibodies are evaluated to determine their binding kinetics. The advantages and disadvantages of the individual approach will be investigated and discussed in detail. In summary, we will raise awareness on how to evaluate and judge results from BIA by using different approaches rather than having to rely on “black box” closed (commercial) software packages.
Collapse
Affiliation(s)
- Monika Conrad
- Institute of Physical and Theoretical Chemistry (IPTC), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 18, 72076, Tübingen, Germany.
| | - Peter Fechner
- Institute of Physical and Theoretical Chemistry (IPTC), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 18, 72076, Tübingen, Germany
| | - Günther Proll
- Institute of Physical and Theoretical Chemistry (IPTC), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 18, 72076, Tübingen, Germany
| | - Günter Gauglitz
- Institute of Physical and Theoretical Chemistry (IPTC), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 18, 72076, Tübingen, Germany
| |
Collapse
|
17
|
Banerjee A, Li D, Guo Y, Mahgoub B, Paragas L, Slobin J, Mei Z, Manafi A, Hata A, Li K, Shi L, Westwick J, Slingluff C, Lazear E, Krupnick AS. Retargeting IL-2 Signaling to NKG2D-Expressing Tumor-Infiltrating Leukocytes Improves Adoptive Transfer Immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:333-343. [PMID: 34155069 PMCID: PMC8688582 DOI: 10.4049/jimmunol.2000926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 04/28/2021] [Indexed: 11/19/2022]
Abstract
Ex vivo expansion followed by reinfusion of tumor-infiltrating leukocytes (TILs) has been used successfully for the treatment of multiple malignancies. Most protocols rely on the use of the cytokine IL-2 to expand TILs prior to reinfusion. In addition, TIL administration relies on systemic administration of IL-2 after reinfusion to support transferred cell survival. The use of IL-2, however, can be problematic because of its preferential expansion of regulatory T and myeloid cells as well as its systemic side effects. In this study, we describe the use of a novel IL-2 mutant retargeted to NKG2D rather than the high-affinity IL-2R for TIL-mediated immunotherapy in a murine model of malignant melanoma. We demonstrate that the NKG2D-retargeted IL-2 (called OMCPmutIL-2) preferentially expands TIL-resident CTLs, such as CD8+ T cells, NK cells, and γδT cells, whereas wild-type IL-2 provides a growth advantage for CD4+Foxp3+ T cells as well as myeloid cells. OMCPmutIL-2-expanded CTLs express higher levels of tumor-homing receptors, such as LFA-1, CD49a, and CXCR3, which correlate with TIL localization to the tumor bed after i.v. injection. Consistent with this, OMCPmutIL-2-expanded TILs provided superior tumor control compared with those expanded in wild-type IL-2. Our data demonstrate that adoptive transfer immunotherapy can be improved by rational retargeting of cytokine signaling to NKG2D-expressing CTLs rather than indiscriminate expansion of all TILs.
Collapse
Affiliation(s)
- Anirban Banerjee
- Department of Surgery, University of Virginia, Charlottesville, VA;
| | - Dongge Li
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Yizhan Guo
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Bayan Mahgoub
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Lea Paragas
- Department of Surgery, University of Virginia, Charlottesville, VA
| | | | - Zhongcheng Mei
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Amir Manafi
- Department of Surgery, University of Virginia, Charlottesville, VA
| | - Atsushi Hata
- Department of Surgery, University of Virginia, Charlottesville, VA
- Department of General Thoracic Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kang Li
- The First Affiliated Hospital of Xi'an, Jiaotong University, Shaanxi, China; and
| | - Lei Shi
- The First Affiliated Hospital of Xi'an, Jiaotong University, Shaanxi, China; and
| | | | - Craig Slingluff
- Department of Surgery, University of Virginia, Charlottesville, VA
| | | | - Alexander Sasha Krupnick
- Department of Surgery, University of Virginia, Charlottesville, VA;
- Courier Therapeutics, Houston, TX
| |
Collapse
|
18
|
Freitas FC, Ferreira PHB, Favaro DC, Oliveira RJD. Shedding Light on the Inhibitory Mechanisms of SARS-CoV-1/CoV-2 Spike Proteins by ACE2-Designed Peptides. J Chem Inf Model 2021; 61:1226-1243. [PMID: 33619962 PMCID: PMC7931628 DOI: 10.1021/acs.jcim.0c01320] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Indexed: 01/07/2023]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the host cellular receptor that locks onto the surface spike protein of the 2002 SARS coronavirus (SARS-CoV-1) and of the novel, highly transmissible and deadly 2019 SARS-CoV-2, responsible for the COVID-19 pandemic. One strategy to avoid the virus infection is to design peptides by extracting the human ACE2 peptidase domain α1-helix, which would bind to the coronavirus surface protein, preventing the virus entry into the host cells. The natural α1-helix peptide has a stronger affinity to SARS-CoV-2 than to SARS-CoV-1. Another peptide was designed by joining α1 with the second portion of ACE2 that is far in the peptidase sequence yet grafted in the spike protein interface with ACE2. Previous studies have shown that, among several α1-based peptides, the hybrid peptidic scaffold is the one with the highest/strongest affinity for SARS-CoV-1, which is comparable to the full-length ACE2 affinity. In this work, binding and folding dynamics of the natural and designed ACE2-based peptides were simulated by the well-known coarse-grained structure-based model, with the computed thermodynamic quantities correlating with the experimental binding affinity data. Furthermore, theoretical kinetic analysis of native contact formation revealed the distinction between these processes in the presence of the different binding partners SARS-CoV-1 and SARS-CoV-2 spike domains. Additionally, our results indicate the existence of a two-state folding mechanism for the designed peptide en route to bind to the spike proteins, in contrast to a downhill mechanism for the natural α1-helix peptides. The presented low-cost simulation protocol demonstrated its efficiency in evaluating binding affinities and identifying the mechanisms involved in the neutralization of spike-ACE2 interaction by designed peptides. Finally, the protocol can be used as a computer-based screening of more potent designed peptides by experimentalists searching for new therapeutics against COVID-19.
Collapse
Affiliation(s)
- Frederico Campos Freitas
- Laboratório de Biofísica Teórica,
Departamento de Física, Instituto de Ciências Exatas, Naturais
e Educação, Universidade Federal do Triângulo
Mineiro, Uberaba, MG 38064-200, Brazil
| | - Paulo Henrique Borges Ferreira
- Laboratório de Biofísica Teórica,
Departamento de Física, Instituto de Ciências Exatas, Naturais
e Educação, Universidade Federal do Triângulo
Mineiro, Uberaba, MG 38064-200, Brazil
| | - Denize Cristina Favaro
- Departamento de Química Orgânica,
Instituto de Química, Universidade Estadual de
Campinas, São Paulo, SP 13083-970, Brazil
| | - Ronaldo Junio de Oliveira
- Laboratório de Biofísica Teórica,
Departamento de Física, Instituto de Ciências Exatas, Naturais
e Educação, Universidade Federal do Triângulo
Mineiro, Uberaba, MG 38064-200, Brazil
| |
Collapse
|
19
|
Zhang Y, Zhang M, Liu Y, Zhang D, Tang Y, Ren B, Zheng J. Dual amyloid cross-seeding reveals steric zipper-facilitated fibrillization and pathological links between protein misfolding diseases. J Mater Chem B 2021; 9:3300-3316. [PMID: 33651875 DOI: 10.1039/d0tb02958k] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Amyloid cross-seeding, as a result of direct interaction and co-aggregation between different disease-causative peptides, is considered as a main mechanism for the spread of the overlapping pathology across different cells and tissues between different protein-misfolding diseases (PMDs). Despite the biomedical significance of amyloid cross-seeding in amyloidogenesis, it remains a great challenge to discover amyloid cross-seeding systems and reveal their cross-seeding structures and mechanisms. Herein, we are the first to report that GNNQQNY - a short fragment from yeast prion protein Sup35 - can cross-seed with both amyloid-β (Aβ, associated with Alzheimer's disease) and human islet amyloid polypeptide (hIAPP, associated with type II diabetes) to form β-structure-rich assemblies and to accelerate amyloid fibrillization. Dry, steric β-zippers, formed by the two β-sheets of different amyloid peptides, provide generally interactive and structural motifs to facilitate amyloid cross-seeding. The presence of different steric β-zippers in a variety of GNNQQNY-Aβ and GNNQQNY-hIAPP assemblies also explains amyloid polymorphism. In addition, alteration of steric zipper formation by single-point mutations of GNNQQNY and interactions of GNNQQNY with different Aβ and hIAPP seeds leads to different amyloid cross-seeding efficiencies, further confirming the existence of cross-seeding barriers. This work offers a better structural-based understanding of amyloid cross-seeding mechanisms linked to different PMDs.
Collapse
Affiliation(s)
- Yanxian Zhang
- Department of Chemical, Biomolecular, and Corrosion Engineering The University of Akron, Ohio, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
Mihoc D, Lupu LM, Wiegand P, Kleinekofort W, Müller O, Völklein F, Glocker MO, Barka F, Barka G, Przybylski M. Antibody Epitope and Affinity Determination of the Myocardial Infarction Marker Myoglobin by SPR-Biosensor Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:106-113. [PMID: 32838528 DOI: 10.1021/jasms.0c00234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Myoglobin (MG) is a biomarker for heart muscle injury, making it a potential target protein for early detection of myocardial infarction. Elevated myoglobin levels alone have low specificity for acute myocardial infarction (AMI) but in combination with cardiac troponin T have been considered highly efficient diagnostic biomarkers. Myoglobin is a monomeric heme protein with a molecular weight of 17 kDa that is found in skeletal and cardiac tissue as an intracellular storage unit of oxygen. MG consists of eight α-helices connected by loops and a heme group responsible for oxygen-binding. Monoclonal antibodies are widely used analytical tools in biomedical research and have been employed for immunoanalytical detection of MG. However, the epitope(s) recognized by MG antibodies have been hitherto unknown. Precise molecular identification of the epitope(s) recognized by antibodies is of key importance for the development of MG as a diagnostic biomarker. The epitope of a monoclonal MG antibody was identified by proteolytic epitope extraction mass spectrometry in combination with surface plasmon resonance (SPR) biosensor analysis. The MG antibody was immobilized both on an affinity microcolumn and a gold SPR chip. The SPR kinetic analysis provided an affinity-binding constant KD of 270 nM for MG. Binding of a tryptic peptide mixture followed by elution of the epitope from the SPR-MS affinity interface by mild acidification provided a single-epitope peptide located at the C-terminus [146-153] [YKELGFQG] of MG. The specificity and affinity of the epitope were ascertained by synthesis and affinity-mass spectrometric characterization of the epitope peptide.
Collapse
Affiliation(s)
- Delia Mihoc
- Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry (STZ), Marktstrasse 29, 65428 Rüsselsheim am Main, Germany
| | - Loredana-Mirela Lupu
- Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry (STZ), Marktstrasse 29, 65428 Rüsselsheim am Main, Germany
| | - Pascal Wiegand
- Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry (STZ), Marktstrasse 29, 65428 Rüsselsheim am Main, Germany
| | - Wolfgang Kleinekofort
- Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry (STZ), Marktstrasse 29, 65428 Rüsselsheim am Main, Germany
- Institute for Microtechnologies (IMTECH), Rhein Main University, 65428 Rüsselsheim am Main, Germany
| | - Oliver Müller
- Institute for Microtechnologies (IMTECH), Rhein Main University, 65428 Rüsselsheim am Main, Germany
| | - Friedemann Völklein
- Institute for Microtechnologies (IMTECH), Rhein Main University, 65428 Rüsselsheim am Main, Germany
| | - Michael O Glocker
- Department of Immunology, Proteome Centre, Medical University Rostock, Schillingallee 69, 18055 Rostock, Germany
| | - Frederik Barka
- Sunchrom GmbH, Industriestr. 27, 61381 Friedrichsdorf, Germany
| | - Günes Barka
- Sunchrom GmbH, Industriestr. 27, 61381 Friedrichsdorf, Germany
| | - Michael Przybylski
- Steinbeis Transfer Centre for Biopolymer Analysis and Biomedical Mass Spectrometry (STZ), Marktstrasse 29, 65428 Rüsselsheim am Main, Germany
| |
Collapse
|
21
|
Hernandez Alvarez B, Skokowa J, Coles M, Mir P, Nasri M, Maksymenko K, Weidmann L, Rogers KW, Welte K, Lupas AN, Müller P, ElGamacy M. Design of novel granulopoietic proteins by topological rescaffolding. PLoS Biol 2020; 18:e3000919. [PMID: 33351791 PMCID: PMC7755208 DOI: 10.1371/journal.pbio.3000919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 11/24/2020] [Indexed: 11/18/2022] Open
Abstract
Computational protein design is rapidly becoming more powerful, and improving the accuracy of computational methods would greatly streamline protein engineering by eliminating the need for empirical optimization in the laboratory. In this work, we set out to design novel granulopoietic agents using a rescaffolding strategy with the goal of achieving simpler and more stable proteins. All of the 4 experimentally tested designs were folded, monomeric, and stable, while the 2 determined structures agreed with the design models within less than 2.5 Å. Despite the lack of significant topological or sequence similarity to their natural granulopoietic counterpart, 2 designs bound to the granulocyte colony-stimulating factor (G-CSF) receptor and exhibited potent, but delayed, in vitro proliferative activity in a G-CSF-dependent cell line. Interestingly, the designs also induced proliferation and differentiation of primary human hematopoietic stem cells into mature granulocytes, highlighting the utility of our approach to develop highly active therapeutic leads purely based on computational design. De novo designed cytokines that activate the G-CSF receptor show that the receptor-binding information can be encoded onto stable, miniaturised protein scaffolds that possess potent granulopoietic activity; such novel proteins provide for ideal candidates for protein-based therapeutics.
Collapse
Affiliation(s)
| | - Julia Skokowa
- University Hospital Tübingen, Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Germany
- * E-mail: (JS); (ME)
| | - Murray Coles
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Perihan Mir
- University Hospital Tübingen, Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Germany
| | - Masoud Nasri
- University Hospital Tübingen, Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Germany
| | | | - Laura Weidmann
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | | | - Karl Welte
- University Hospital Tübingen, Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Germany
| | - Andrei N. Lupas
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Patrick Müller
- University Hospital Tübingen, Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Germany
- Friedrich Miescher Laboratory of the Max Planck Society Tübingen, Germany
| | - Mohammad ElGamacy
- Max Planck Institute for Developmental Biology, Tübingen, Germany
- University Hospital Tübingen, Division of Translational Oncology, Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Germany
- Friedrich Miescher Laboratory of the Max Planck Society Tübingen, Germany
- Heliopolis Biotechnology Ltd., London, United Kingdom
- * E-mail: (JS); (ME)
| |
Collapse
|
22
|
Suire CN, Abdul-Hay SO, Sahara T, Kang D, Brizuela MK, Saftig P, Dickson DW, Rosenberry TL, Leissring MA. Cathepsin D regulates cerebral Aβ42/40 ratios via differential degradation of Aβ42 and Aβ40. ALZHEIMERS RESEARCH & THERAPY 2020; 12:80. [PMID: 32631408 PMCID: PMC7339583 DOI: 10.1186/s13195-020-00649-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/24/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cathepsin D (CatD) is a lysosomal protease that degrades both the amyloid β-protein (Aβ) and the microtubule-associated protein, tau, and has been genetically linked to late-onset Alzheimer disease (AD). Here, we sought to examine the consequences of genetic deletion of CatD on Aβ proteostasis in vivo and to more completely characterize the degradation of Aβ42 and Aβ40 by CatD. METHODS We quantified Aβ degradation rates and levels of endogenous Aβ42 and Aβ40 in the brains of CatD-null (CatD-KO), heterozygous null (CatD-HET), and wild-type (WT) control mice. CatD-KO mice die by ~ 4 weeks of age, so tissues from younger mice, as well as embryonic neuronal cultures, were investigated. Enzymological assays and surface plasmon resonance were employed to quantify the kinetic parameters (KM, kcat) of CatD-mediated degradation of monomeric human Aβ42 vs. Aβ40, and the degradation of aggregated Aβ42 species was also characterized. Competitive inhibition assays were used to interrogate the relative inhibition of full-length human and mouse Aβ42 and Aβ40, as well as corresponding p3 fragments. RESULTS Genetic deletion of CatD resulted in 3- to 4-fold increases in insoluble, endogenous cerebral Aβ42 and Aβ40, exceeding the increases produced by deletion of an insulin-degrading enzyme, neprilysin or both, together with readily detectable intralysosomal deposits of endogenous Aβ42-all by 3 weeks of age. Quite significantly, CatD-KO mice exhibited ~ 30% increases in Aβ42/40 ratios, comparable to those induced by presenilin mutations. Mechanistically, the perturbed Aβ42/40 ratios were attributable to pronounced differences in the kinetics of degradation of Aβ42 vis-à-vis Aβ40. Specifically, Aβ42 shows a low-nanomolar affinity for CatD, along with an exceptionally slow turnover rate that, together, renders Aβ42 a highly potent competitive inhibitor of CatD. Notably, the marked differences in the processing of Aβ42 vs. Aβ40 also extend to p3 fragments ending at positions 42 vs. 40. CONCLUSIONS Our findings identify CatD as the principal intracellular Aβ-degrading protease identified to date, one that regulates Aβ42/40 ratios via differential degradation of Aβ42 vs. Aβ40. The finding that Aβ42 is a potent competitive inhibitor of CatD suggests a possible mechanistic link between elevations in Aβ42 and downstream pathological sequelae in AD.
Collapse
Affiliation(s)
- Caitlin N Suire
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA.,Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92697, USA
| | - Samer O Abdul-Hay
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Tomoko Sahara
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Dongcheul Kang
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Monica K Brizuela
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA
| | - Paul Saftig
- Institut für Biochemie, Christian-Albrechts-Universität zu Kiel, 24098, Kiel, Germany
| | - Dennis W Dickson
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | | | - Malcolm A Leissring
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, 92697, USA. .,Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, 32224, USA.
| |
Collapse
|
23
|
Gauglitz G. Critical assessment of relevant methods in the field of biosensors with direct optical detection based on fibers and waveguides using plasmonic, resonance, and interference effects. Anal Bioanal Chem 2020; 412:3317-3349. [PMID: 32313998 PMCID: PMC7214504 DOI: 10.1007/s00216-020-02581-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/16/2022]
Abstract
Direct optical detection has proven to be a highly interesting tool in biomolecular interaction analysis to be used in drug discovery, ligand/receptor interactions, environmental analysis, clinical diagnostics, screening of large data volumes in immunology, cancer therapy, or personalized medicine. In this review, the fundamental optical principles and applications are reviewed. Devices are based on concepts such as refractometry, evanescent field, waveguides modes, reflectometry, resonance and/or interference. They are realized in ring resonators; prism couplers; surface plasmon resonance; resonant mirror; Bragg grating; grating couplers; photonic crystals, Mach-Zehnder, Young, Hartman interferometers; backscattering; ellipsometry; or reflectance interferometry. The physical theories of various optical principles have already been reviewed in detail elsewhere and are therefore only cited. This review provides an overall survey on the application of these methods in direct optical biosensing. The "historical" development of the main principles is given to understand the various, and sometimes only slightly modified variations published as "new" methods or the use of a new acronym and commercialization by different companies. Improvement of optics is only one way to increase the quality of biosensors. Additional essential aspects are the surface modification of transducers, immobilization strategies, selection of recognition elements, the influence of non-specific interaction, selectivity, and sensitivity. Furthermore, papers use for reporting minimal amounts of detectable analyte terms such as value of mass, moles, grams, or mol/L which are difficult to compare. Both these essential aspects (i.e., biochemistry and the presentation of LOD values) can be discussed only in brief (but references are provided) in order to prevent the paper from becoming too long. The review will concentrate on a comparison of the optical methods, their application, and the resulting bioanalytical quality.
Collapse
Affiliation(s)
- Günter Gauglitz
- Institute of Physical and Theoretical Chemistry, Eberhard Karls Universität, Auf der Morgenstelle 18, 72076, Tübingen, Germany.
| |
Collapse
|
24
|
Norval LW, Krämer SD, Gao M, Herz T, Li J, Rath C, Wöhrle J, Günther S, Roth G. KOFFI and Anabel 2.0-a new binding kinetics database and its integration in an open-source binding analysis software. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2020; 2019:5585575. [PMID: 31608948 PMCID: PMC6790968 DOI: 10.1093/database/baz101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 12/31/2022]
Abstract
The kinetics of featured interactions (KOFFI) database is a novel tool and resource for binding kinetics data from biomolecular interactions. While binding kinetics data are abundant in literature, finding valuable information is a laborious task. We used text extraction methods to store binding rates (association, dissociation) as well as corresponding meta-information (e.g. methods, devices) in a novel database. To date, over 270 articles were manually curated and binding data on over 1705 interactions was collected and stored in the (KOFFI) database. Moreover, the KOFFI database application programming interface was implemented in Anabel (open-source software for the analysis of binding interactions), enabling users to directly compare their own binding data analyses with related experiments described in the database.
Collapse
Affiliation(s)
- Leo William Norval
- ZBSA Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse 49, D-79104 Freiburg, Germany.,Institute of Pharmaceutical Sciences, Pharmaceutical Bioinformatics, Albert-Ludwigs-University Freiburg, Hermann-Herder-Straße 9, D-79104 Freiburg, Germany
| | - Stefan Daniel Krämer
- ZBSA Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse 49, D-79104 Freiburg, Germany.,Faculty for Biology, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Mingjie Gao
- Institute of Pharmaceutical Sciences, Pharmaceutical Bioinformatics, Albert-Ludwigs-University Freiburg, Hermann-Herder-Straße 9, D-79104 Freiburg, Germany
| | - Tobias Herz
- ZBSA Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse 49, D-79104 Freiburg, Germany.,Faculty for Biology, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jianyu Li
- Institute of Pharmaceutical Sciences, Pharmaceutical Bioinformatics, Albert-Ludwigs-University Freiburg, Hermann-Herder-Straße 9, D-79104 Freiburg, Germany
| | - Christin Rath
- ZBSA Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse 49, D-79104 Freiburg, Germany.,Faculty for Biology, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany.,BioCopy GmbH, Spechtweg 25, D-79110 Freiburg, Germany.,BIOSS Center for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestrasse 18, D-79104 Freiburg, Germany
| | - Johannes Wöhrle
- ZBSA Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse 49, D-79104 Freiburg, Germany.,IMTEK Department of Microsystems Engineering, Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 103, D-79110 Freiburg, Germany
| | - Stefan Günther
- Institute of Pharmaceutical Sciences, Pharmaceutical Bioinformatics, Albert-Ludwigs-University Freiburg, Hermann-Herder-Straße 9, D-79104 Freiburg, Germany
| | - Günter Roth
- ZBSA Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse 49, D-79104 Freiburg, Germany.,Faculty for Biology, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany.,BioCopy GmbH, Spechtweg 25, D-79110 Freiburg, Germany.,BIOSS Center for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schänzlestrasse 18, D-79104 Freiburg, Germany
| |
Collapse
|
25
|
Krämer SD, Wöhrle J, Meyer PA, Urban GA, Roth G. How to copy and paste DNA microarrays. Sci Rep 2019; 9:13940. [PMID: 31558745 PMCID: PMC6763488 DOI: 10.1038/s41598-019-50371-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/11/2019] [Indexed: 12/31/2022] Open
Abstract
Analogous to a photocopier, we developed a DNA microarray copy technique and were able to copy patterned original DNA microarrays. With this process the appearance of the copied DNA microarray can also be altered compared to the original by producing copies of different resolutions. As a homage to the very first photocopy made by Chester Charlson and Otto Kornei, we performed a lookalike DNA microarray copy exactly 80 years later. Those copies were also used for label-free real-time kinetic binding assays of apo-dCas9 to double stranded DNA and of thrombin to single stranded DNA. Since each DNA microarray copy was made with only 5 µl of spPCR mix, the whole process is cost-efficient. Hence, our DNA microarray copier has a great potential for becoming a standard lab tool.
Collapse
Affiliation(s)
- Stefan D Krämer
- ZBSA - Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse. 49, D-79104, Freiburg, Germany. .,Faculty for Biology, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 1, D-79104, Freiburg, Germany.
| | - Johannes Wöhrle
- ZBSA - Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse. 49, D-79104, Freiburg, Germany.,IMTEK - Dep. of Microsystems Engineering, Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 103, D-79110, Freiburg, Germany
| | - Philipp A Meyer
- ZBSA - Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse. 49, D-79104, Freiburg, Germany.,IMTEK - Dep. of Microsystems Engineering, Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 103, D-79110, Freiburg, Germany
| | - Gerald A Urban
- IMTEK - Dep. of Microsystems Engineering, Albert-Ludwigs-University Freiburg, Georges-Köhler-Allee 103, D-79110, Freiburg, Germany.,BIOSS - Center for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 18, D-79104, Freiburg, Germany
| | - Günter Roth
- ZBSA - Center for Biological Systems Analysis, Albert-Ludwigs-University Freiburg, Habsburgerstrasse. 49, D-79104, Freiburg, Germany.,Faculty for Biology, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 1, D-79104, Freiburg, Germany.,BioCopy GmbH, Spechtweg 25, D-79110, Freiburg, Germany.,BIOSS - Center for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 18, D-79104, Freiburg, Germany.,BioCopy Holding AG, Industriestrasse 15, 8355, Aadorf, Switzerland
| |
Collapse
|
26
|
Ito S, Saito M, Yoshida M, Takeuchi K, Doi T, Nagata K. A BRET-based assay reveals collagen-Hsp47 interaction dynamics in the endoplasmic reticulum and small-molecule inhibition of this interaction. J Biol Chem 2019; 294:15962-15972. [PMID: 31492754 PMCID: PMC6827286 DOI: 10.1074/jbc.ra119.010567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 08/23/2019] [Indexed: 01/01/2023] Open
Abstract
Molecular chaperones perform pivotal roles in proteostasis by engaging in protein–protein interactions (PPIs). The collagen-specific molecular chaperone Hsp47 (heat shock protein 47) interacts with procollagen in the endoplasmic reticulum (ER) and plays crucial roles in collagen synthesis. PPIs between Hsp47 and collagen could offer a therapeutic target for fibrosis, which is characterized by abnormal collagen accumulation in the extracellular matrix of fibrotic organs. Herein, we established a bioluminescence resonance energy transfer (BRET) system for assessing Hsp47–collagen interaction dynamics within the ER. After optimization and validation of the method, we could demonstrate inhibition of the interaction between Hsp47 and collagen by a small molecule (Col003) in the ER. Using the BRET system, we also found that Hsp47 interacts not only with the Gly-Pro-Arg motif but also weakly with Gly-Pro-Hyp motifs of triple-helical collagen in cells. Moreover, we found that the serpin loop of Hsp47 (SerpinH1) contributes to its binding to collagen. We propose that the method developed here can provide valuable information on PPIs between Hsp47 and collagen and on the effects of PPI inhibitors important for the management of fibrotic disorders.
Collapse
Affiliation(s)
- Shinya Ito
- Institute for Protein Dynamics, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Masazumi Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Masahito Yoshida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Koh Takeuchi
- National Institute of Advanced Industrial Science and Technology, Tokyo 135-0064, Japan
| | - Takayuki Doi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai 980-8578, Japan
| | - Kazuhiro Nagata
- Institute for Protein Dynamics, Kyoto Sangyo University, Kyoto 603-8555, Japan .,Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan.,CREST, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto 603-8555, Japan
| |
Collapse
|
27
|
Rath C, Burger J, Norval L, Kraemer SD, Gensch N, van der Kooi A, Reinemann C, O'Sullivan C, Svobodova M, Roth G. Comparison of different label-free imaging high-throughput biosensing systems for aptamer binding measurements using thrombin aptamers. Anal Biochem 2019; 583:113323. [PMID: 31129134 DOI: 10.1016/j.ab.2019.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/10/2019] [Accepted: 05/16/2019] [Indexed: 01/18/2023]
Abstract
To enable the analysis of several hundreds to thousands of interactions in parallel, high-throughput systems were developed. We used established thrombin aptamer assays to compare three such high-throughput imaging systems as well as analysis software and user influence. In addition to our own iRIf-system, we applied bscreen and IBIS-MX96. As non-imaging reference systems we used Octet-RED96, Biacore3000, and Monolith-NT.115. In this study we measured 1378 data points. Our results show that all systems are suitable for analyzing binding kinetics, but the kinetic constants as well as the ranking of the selected aptamers depend significantly on the applied system and user. We provide an insight into the signal generation principles, the systems and the results generated for thrombin aptamers. It should contribute to the awareness that binding constants cannot be determined as easily as other constants. Since many parameters like surface chemistry, biosensor type and buffer composition may change binding behavior, the experimenter should be aware that a system and assay dependent KD is determined. Frequently, certain conditions that are best suited for a given biosensing system cannot be transferred to other systems. Therefore, we strongly recommend using at least two different systems in parallel to achieve meaningful results.
Collapse
Affiliation(s)
- Christin Rath
- Laboratory for Microarray Copying, Center for Biological Systems Analysis (ZBSA), University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany; Faculty for Biology, Biology 3, University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany; Centre for Biological Signaling Studies (BIOSS), University of Freiburg, 79104, Freiburg, Germany; BioCopy GmbH, 79110 Freiburg, Germany.
| | - Juergen Burger
- Laboratory for Microarray Copying, Center for Biological Systems Analysis (ZBSA), University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany; BioCopy GmbH, 79110 Freiburg, Germany.
| | - Leo Norval
- Laboratory for Microarray Copying, Center for Biological Systems Analysis (ZBSA), University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany.
| | - Stefan Daniel Kraemer
- Laboratory for Microarray Copying, Center for Biological Systems Analysis (ZBSA), University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany; Faculty for Biology, Biology 3, University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany.
| | - Nicole Gensch
- Core Facility Signalling Factory, Centre for Biological Signaling Studies (BIOSS), University of Freiburg, 79104 Freiburg, Germany.
| | | | - Christine Reinemann
- Helmholtz Centre for Environmental Research GmbH (UFZ), Permoserstr. 15, 04318, Leipzig, Germany.
| | - Ciara O'Sullivan
- Departament d'Enginyería Química, Universitat Rovira i Virgili, 43007, Tarragona, Spain; Institució Catalana de Recerca i Estudis Avançats, 08010, Barcelona, Spain.
| | - Marketa Svobodova
- Departament d'Enginyería Química, Universitat Rovira i Virgili, 43007, Tarragona, Spain.
| | - Guenter Roth
- Laboratory for Microarray Copying, Center for Biological Systems Analysis (ZBSA), University of Freiburg, Habsburgerstrasse 49, 79104, Freiburg, Germany; Faculty for Biology, Biology 3, University of Freiburg, Schänzlestrasse 1, 79104, Freiburg, Germany; Centre for Biological Signaling Studies (BIOSS), University of Freiburg, 79104, Freiburg, Germany; BioCopy GmbH, 79110 Freiburg, Germany.
| |
Collapse
|