1
|
Mengrani Z, Hong W, Palma M. DNA-Mediated Carbon Nanotubes Heterojunction Assembly. ACS NANOSCIENCE AU 2024; 4:391-398. [PMID: 39713723 PMCID: PMC11659895 DOI: 10.1021/acsnanoscienceau.4c00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 12/24/2024]
Abstract
Herein, we present a strategy for the controlled assembly of single-walled carbon nanotube (SWCNT) linear junctions mediated by DNA as a functional linker. We demonstrate this by employing SWCNTs of two different chiralities via the specific design of DNA sequences and chiral selection. Streptavidin and AuNP labeling of the SWCNT sidewalls demonstrate the presence of two different chirality within each individual CNT-DNA-CNT junction. These one-dimensional nanohybrids were further organized from solution to devices. The approach we developed is of general applicability for the assembly of functional nanohybrids based on carbon nanotubes toward functional applications.
Collapse
Affiliation(s)
| | | | - Matteo Palma
- Department of Chemistry, Queen Mary University of London, London E1 4NS, U.K.
| |
Collapse
|
2
|
Gu X, He L, Zhang J, Xu H, Shen H, Huang R, Li Z. Recent Advances in Wash-Free Detection Methods of Extracellular Vesicles: A Mini Review. ACS Sens 2024; 9:5626-5641. [PMID: 39446112 DOI: 10.1021/acssensors.4c00315] [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] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs) are emerging biomarkers in liquid biopsy that have gained increasing attention in disease diagnosis and prognosis monitoring. Most reported detection methods require the isolation of EVs from complex body liquids, often involving multiple washing steps to remove excess reagents and eliminate background interference. Nonetheless, these methods not only cause the loss of EVs but also result in poor repeatability and prolonged detection duration. The focus on wash-free detection methods is increasing due to the specific ability to avoid the removal of surplus reagents and, in some cases, even the isolation and purification of EVs. Viewing from different methodological perspectives, this review summarizes the recent advances in wash-free detection of EVs, containing aggregation induction, proximity sensing, allosteric probes, phase separation, Roman spectroscopy, field-effect transistor and microcantilever. The pros and cons of each detection strategy are impartially evaluated and this review concludes the prospects for future developments in this field.
Collapse
Affiliation(s)
- Xinrui Gu
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Lei He
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Jinsong Zhang
- Clinical Laboratory, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Hongpan Xu
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Han Shen
- Clinical Laboratory, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| | - Rongrong Huang
- School of Pharmaceutical Sciences, Nanjing Tech University, South Puzhu Road 30, Nanjing, Jiangsu Province 211816, China
| | - Zhiyang Li
- Clinical Laboratory, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Zhongshan Road 321, Nanjing, Jiangsu Province 210008, China
| |
Collapse
|
3
|
Sam G, Chen S, Rehm BHA. Functionalisation of polyhydroxybutyrate for diagnostic uses. N Biotechnol 2024; 85:9-15. [PMID: 39549939 DOI: 10.1016/j.nbt.2024.11.002] [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: 06/28/2024] [Revised: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 11/18/2024]
Abstract
Polyhydroxybutyrate (PHB) is a biodegradable and biocompatible biopolyester, naturally produced and self-assembled as spherical inclusions inside bacteria. These PHB particles contain a hydrophobic PHB core covalently coated with PHB synthase (PhaC), which serves as an anchoring linker for foreign proteins of interest. Protein engineering of PhaC enables the display of biologically active protein functions on the surface of PHB particles suitable for different applications. Many biomolecules, such as e.g. antigens, enzymes, fluorescent proteins were immobilized to PHB particles and exhibited superior functionalities when compared to their respective soluble counterparts. Recently, PHB particles have been successfully applied for various diagnostics applications. This mini review provides an overview of the unique design space of PHB particles towards the development of safe and cost-effective diagnostic tools, and highlights the important research progresses of manufacturing PHB particles-based diagnostics.
Collapse
Affiliation(s)
- Gayathri Sam
- Centre for Cell Factories and Biopolymers (CCFB), Institute for Biomedicine and Glycomics, Griffith University (Nathan Campus), QLD 4111, Australia
| | - Shuxiong Chen
- Centre for Cell Factories and Biopolymers (CCFB), Institute for Biomedicine and Glycomics, Griffith University (Nathan Campus), QLD 4111, Australia.
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers (CCFB), Institute for Biomedicine and Glycomics, Griffith University (Nathan Campus), QLD 4111, Australia; Menzies Health Institute Queensland (MHIQ), Griffith University (Gold Coast Campus), QLD 4215, Australia.
| |
Collapse
|
4
|
Shilova NV, Polyakova SM, Nokel AY, Lipatnikov AD, Gordeeva EA, Lavrenteva MV, Bovin NV. (Strept)avidin Binds Glycoconjugates. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2023-2027. [PMID: 39647829 DOI: 10.1134/s0006297924110142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 12/10/2024]
Abstract
Fluorescently labeled and conjugated (strept)avidins are widely used for imaging biotinylated molecules in immunological assays and histochemistry. We showed that besides biotin, these proteins bind glycans, including fragments of mammalian glycoproteins and glycolipids, in particular, ABO blood group antigens, oligolactosamines, and 6-O-sulfated oligosaccharides. This interaction is inhibited in a dose-dependent manner by micromolar concentrations of polymeric, but not monomeric, glycan conjugates (i.e., requires polyvalence). Taking into account the cluster organization of cell glycans (glycoproteins and glycolipids), the ability of (strept)avidins to bind glycans might be a source of errors in the analysis of carbohydrate-containing samples, which can be prevented by avoiding a large excess of (strept)avidin in analytical systems.
Collapse
Affiliation(s)
- Nadezhda V Shilova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov of the Ministry of Health of the Russian Federation, Moscow, 117997, Russia
| | - Svetlana M Polyakova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Alexey Yu Nokel
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V. I. Kulakov of the Ministry of Health of the Russian Federation, Moscow, 117997, Russia
| | - Alexander D Lipatnikov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Elena A Gordeeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Marina V Lavrenteva
- Biotechnology Department, MIREA - Russian Technological University, Moscow, 119571, Russia
| | - Nicolai V Bovin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| |
Collapse
|
5
|
Salazar Marcano DE, Chen JJ, Moussawi MA, Kalandia G, Anyushin AV, Parac-Vogt TN. Redox-active polyoxovanadates as cofactors in the development of functional protein assemblies. J Inorg Biochem 2024; 260:112687. [PMID: 39142056 DOI: 10.1016/j.jinorgbio.2024.112687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024]
Abstract
The interactions of polyoxovanadates (POVs) with proteins have increasingly attracted interest in recent years due to their potential biomedical applications. This is especially the case because of their redox and catalytic properties, which make them interesting for developing artificial metalloenzymes. Organic-inorganic hybrid hexavanadates in particular offer several advantages over all-inorganic POVs. However, they have been scarcely investigated in biological systems even though, as shown in this work, hybrid hexavanadates are highly stable in aqueous solutions up to relatively high pH. Therefore, a novel bis-biotinylated hexavanadate was synthesized and shown to selectively interact with two biotin-binding proteins, avidin and streptavidin. Bridging interactions between multiple proteins led to their self-assembly into supramolecular bio-inorganic hybrid systems that have potential as artificial enzymes with the hexavanadate core as a redox-active cofactor. Moreover, the structure and charge of the hexavanadate core were determined to enhance the binding affinity and slightly alter the secondary structure of the proteins, which affected the size and speed of formation of the assemblies. Hence, tuning the polyoxometalate (POM) core of hybrid POMs (HPOMs) with protein-binding ligands has been demonstrated to be a potential strategy for controlling the self-assembly process while also enabling the formation of novel POM-based biomaterials that could be of interest in biomedicine.
Collapse
Affiliation(s)
| | - Jieh-Jang Chen
- Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Mhamad Aly Moussawi
- Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | - Givi Kalandia
- Department of Chemistry, KU Leuven, Celestijnenlaan 200F, 3001 Leuven, Belgium
| | | | | |
Collapse
|
6
|
Wang Z, Dai J, He H, Si T, Ng K, Zheng S, Zhou X, Zhou Z, Yuan H, Yang M. Cellulose Nanofibrils of High Immunoaffinity for Efficient Enrichment of Small Extracellular Vesicles. SMALL METHODS 2024; 8:e2400426. [PMID: 38678531 PMCID: PMC11579556 DOI: 10.1002/smtd.202400426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Indexed: 05/01/2024]
Abstract
Extracellular vesicles (EVs), crucial in facilitating the transport of diverse molecular cargoes for intercellular communication, have shown great potential in diagnostics, therapeutics, and drug delivery. The challenge of developing effective preparation methods for EVs is heightened by their intrinsic heterogeneity and complexity. Here, a novel strategy for high EV enrichment is developed by utilizing EV-affinitive-modified cellulose nanofibrils. Specifically, modified cellulose with rich carboxyl groups has outstanding dispersing properties, able to be dispersed into cellulose nanofibrils in solution. These cellulose nanofibrils are utilized as scaffolds for the immobilization of EV-affinitive antibody of CD63 by chemical conjugation. The CD63-modified nanofibrils demonstrate a superior EV capture efficiency of 86.4% compared with other reported methods. The high performance of this system is further validated by the efficient capture of EVs from biological blood plasma, allowing the detection of bioactive markers from EV-derived miRNAs and proteins. The authors envision that these modified cellulose nanofibrils of enhanced capability on EV enrichment will open new avenues in various biomedical applications.
Collapse
Affiliation(s)
- Zesheng Wang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000P. R. China
- Department of Biomedical Sciences, and Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Jun Dai
- Department of Biomedical Sciences, and Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Huimin He
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000P. R. China
- Department of Biomedical Sciences, and Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Tongxu Si
- Department of Biomedical Sciences, and Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Kaki Ng
- Department of Biomedical Sciences, and Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
| | - Shuang Zheng
- Department of Biomedical Sciences, and Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Present address:
Department of Civil EngineeringUniversity of Hong KongPokfulamHong KongP. R. China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000P. R. China
- Department of Biomedical Sciences, and Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| | - Zhihang Zhou
- Department of Gastroenterologythe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Huijun Yuan
- Department of Biochip CenterWuwei Tumor Hospital of Gansu ProvinceGansu730000P. R. China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic TechnologyCity University of Hong Kong Shenzhen Futian Research InstituteShenzhen518000P. R. China
- Department of Biomedical Sciences, and Tung Biomedical Sciences CentreCity University of Hong KongHong Kong999077P. R. China
- Key Laboratory of Biochip TechnologyBiotech and Health CentreShenzhen Research Institute of City University of Hong KongShenzhen518057P. R. China
| |
Collapse
|
7
|
Chen J, Zhang G, Xiao X, Liu D, Peng J, Xiong Y, Lai W. Bifunctional bovine serum albumin modification driven sensitivity-enhanced lateral flow immunoassay for small molecule hazards monitoring in food. Int J Biol Macromol 2024; 282:136915. [PMID: 39476895 DOI: 10.1016/j.ijbiomac.2024.136915] [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: 08/28/2024] [Revised: 09/30/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Traditional lateral flow immunoassays (TLFIAs) are valued for their simplicity, speed, and user-friendliness. However, the specificity of conventional test strips often necessitates large quantities of antigen-protein conjugates for target detection, which can be resource-intensive. Here, we present a strategy aimed at enhancing the universality of test strips while reducing the consumption of antigen-protein conjugates, without compromising sensitivity. By coating streptavidin on the test line and employing bifunctional antigen-protein conjugates (competitor to target and immunoprobe linker), the test strip was thus served as a universal module. We developed three universal lateral flow immunoassays (ULFIAs) for the detection of aflatoxin B1 (AFB1), carbendazim (CBZ), and enrofloxacin (ENR). Compared to traditional methods based on the same aggregation-induced emission fluorescent microspheres, the proposed ULFIAs demonstrated a significant increase in sensitivity, with enhancements of 11.0-fold for AFB1, 10.9-fold for CBZ, and 4.1-fold for ENR. Additionally, this approach substantially reduced the consumption of antigen-protein conjugates by 19.1-fold, 40.9-fold, and 23.8-fold, respectively, thereby promoting greener detection methods. This bifunctional antigen conjugate strategy offers a promising pathway for the sensitive detection of small molecule hazards.
Collapse
Affiliation(s)
- Jiawei Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 East Nanjing Road, Nanchang 330047, China
| | - Gan Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 East Nanjing Road, Nanchang 330047, China
| | - Xiaoyue Xiao
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 East Nanjing Road, Nanchang 330047, China
| | - Daofeng Liu
- Jiangxi Province Key Laboratory of Diagnosing and Tracing of Foodborne Disease, Jiangxi Province Center for Disease Control and Prevention, 555 East Beijing Road, Nanchang 330029, China
| | - Juan Peng
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 East Nanjing Road, Nanchang 330047, China
| | - Yonghua Xiong
- Jiangxi-OAI Joint Research Institute, Nanchang University, Nanchang 330047, China
| | - Weihua Lai
- State Key Laboratory of Food Science and Resources, Nanchang University, 235 East Nanjing Road, Nanchang 330047, China.
| |
Collapse
|
8
|
De Plano LM, Oddo S, Bikard D, Caccamo A, Conoci S. Generation of a Biotin-Tagged Dual-Display Phage. Cells 2024; 13:1696. [PMID: 39451214 PMCID: PMC11506469 DOI: 10.3390/cells13201696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
Phage display is widely used in biomedical research. One of the great advantages of phage display is the specificity of the connection of a foreign peptide exposed outside the capsid to the intended target. Secondary detection systems, which are often laborious and costly, are required to identify and quantify the peptide/target interaction. In this study, we generated a novel dual-display phage to facilitate the detection and quantification of the peptide/target interaction. First, we generated a biotin-tagged phage by adding a small biotin-accepting peptide (sBT) to gene-3 of the M13K07 helper phage. Subsequently, we enhanced the M13K07 biotin-tagged phage by incorporating a selective peptide on gene-8, which is then exposed to the phage capsid. The exposed peptide acts as a probe to bind to a selective molecular target, whose interaction can be readily visualized thanks to the biotinylated phage. Our versatile dual-display phage exhibits high flexibility; by swapping the displayed peptide/probe, one can change the phage target while retaining the sBT gene in-frame with the pIII. We expect the generated biotin-tagged dual phages to be used as a multifunctional probe to couple with several streptavidin-biotin-based systems.
Collapse
Affiliation(s)
- Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Salvatore Oddo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - David Bikard
- Pasteur Institute, University of Paris, Synthetic Biology, 75015 Paris, France
| | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Sabrina Conoci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
- Department of Chemistry G. Ciamician, University of Bologna, Via F. Selmi 2, 40126 Bologna, Italy
- LAB Sense Beyond Nano-DSFTM CNR, Viale F. Stagno d'Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
9
|
Kawashima T, Nakamura M, Sakono M. A one-process production of completely biotinylated proteins in a T7 expression system. Biotechnol Appl Biochem 2024; 71:1070-1078. [PMID: 38770738 DOI: 10.1002/bab.2598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/25/2024] [Indexed: 05/22/2024]
Abstract
Streptavidin is a tetrameric protein with high specificity and affinity for biotin. The interaction between avidin and biotin has become a valuable tool in nanotechnology. In recent years, the site-specific biotin modification of proteins using biotin ligases, such as BirA, has attracted attention. This study established an in vivo method for achieving the complete biotinylation of target proteins using a single plasmid co-expressing BirA and its target proteins. Specifically, a biotin-modified protein was produced in Escherichia coli strain BL21(DE3) using a single plasmid containing genes encoding both BirA and a protein fused to BirA's substrate sequence, Avitag. This approach simplifies the production of biotinylated proteins in E. coli and allows the creation of various biotinylated protein types through gene replacement. Furthermore, the biotin modification rate of the obtained target protein could be evaluated using Native-PAGE without performing complicated isolation operations of biotinylated proteins. In Native-PAGE, biotin-modified proteins and unmodified proteins were confirmed as clearly different bands, and it was possible to easily derive the modification rate from the respective band intensities.
Collapse
Affiliation(s)
- Takuma Kawashima
- Department of Applied Chemistry, Faculty of Engineering, University of Toyama, Toyama, Toyama, Japan
| | - Mitsuki Nakamura
- Department of Applied Chemistry, Faculty of Engineering, University of Toyama, Toyama, Toyama, Japan
| | - Masafumi Sakono
- Department of Applied Chemistry, Faculty of Engineering, University of Toyama, Toyama, Toyama, Japan
| |
Collapse
|
10
|
Prudhomme M, Lakhdar C, Fattaccioli J, Addouche M, Chollet F. Functionalization of microbubbles in a microfluidic chip for biosensing application. Biomed Microdevices 2024; 26:39. [PMID: 39287824 DOI: 10.1007/s10544-024-00721-2] [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] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
Microbubbles are widely used for biomedical applications, ranging from imagery to therapy. In these applications, microbubbles can be functionalized to allow targeted drug delivery or imaging of the human body. However, functionalization of the microbubbles is quite difficult, due to the unstable nature of the gas/liquid interface. In this paper, we describe a simple protocol for rapid functionalization of microbubbles and show how to use them inside a microfluidic chip to develop a novel type of biosensor. The microbubbles are functionalized with biochemical ligand directly at their generation inside the microfluidic chip using a DSPE-PEG-Biotin phospholipid. The microbubbles are then organized inside a chamber before injecting the fluid with the bioanalyte of interest through the static bubbles network. In this proof-of-concept demonstration, we use streptavidin as the bioanalyte of interest. Both functionalization and capture are assessed using fluorescent microscopy thanks to fluorescent labeled chemicals. The main advantages of the proposed technique compared to classical ligand based biosensor using solid surface is its ability to rapidly regenerate the functionalized surface, with the complete functionalization/capture/measurement cycle taking less than 10 min.
Collapse
Affiliation(s)
- Marc Prudhomme
- Institut FEMTO-ST, Université de Franche-Comté, CNRS, Besançon, F-25000, France
| | - Chaimaa Lakhdar
- Institut FEMTO-ST, Université de Franche-Comté, CNRS, Besançon, F-25000, France
| | - Jacques Fattaccioli
- PASTEUR, Département de Chimie, Ecole Normale Supérieure, PSL Université, Sorbonne Université, CNRS, F-75005, Paris, France
- Institut Pierre-Gilles de Gennes pour la Microfluidique, F-75005, Paris, France
| | - Mahmoud Addouche
- Institut FEMTO-ST, Université de Franche-Comté, CNRS, Besançon, F-25000, France
| | - Franck Chollet
- Institut FEMTO-ST, Université de Franche-Comté, CNRS, Besançon, F-25000, France.
| |
Collapse
|
11
|
Lapitan LD, Felisilda BMB, Tiangco CE, Rosin Jose A. Advances in Bioreceptor Layer Engineering in Nanomaterial-based Sensing of Pseudomonas Aeruginosa and its Metabolites. Chem Asian J 2024; 19:e202400090. [PMID: 38781439 DOI: 10.1002/asia.202400090] [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: 01/25/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 05/25/2024]
Abstract
Pseudomonas aeruginosa is a pathogen that infects wounds and burns and causes severe infections in immunocompromised humans. The high virulence, the rise of antibiotic-resistant strains, and the easy transmissibility of P. aeruginosa necessitate its fast detection and control. The gold standard for detecting P. aeruginosa, the plate culture method, though reliable, takes several days to complete. Therefore, developing accurate, rapid, and easy-to-use diagnostic tools for P. aeruginosa is highly desirable. Nanomaterial-based biosensors are at the forefront of detecting P. aeruginosa and its secondary metabolites. This review summarises the biorecognition elements, biomarkers, immobilisation strategies, and current state-of-the-art biosensors for P. aeruginosa. The review highlights the underlying principles of bioreceptor layer engineering and the design of optical, electrochemical, mass-based, and thermal biosensors based on nanomaterials. The advantages and disadvantages of these biosensors and their future point-of-care applications are also discussed. This review outlines significant advancements in biosensors and sensors for detecting P. aeruginosa and its metabolites. Research efforts have identified biorecognition elements specific and selective towards P. aeruginosa. The stability, ease of preparation, cost-effectiveness, and integration of these biorecognition elements onto transducers are pivotal for their application in biosensors and sensors. At the same time, when developing sensors for clinically significant analytes such as P. aeruginosa, virulence factors need to be addressed, such as the sensor's sensitivity, reliability, and response time in samples obtained from patients. The point-of-care applicability of the developed sensor may be an added advantage since it enables onsite determination. In this context, optical methods developed for P. aeruginosa offer promising potential.
Collapse
Affiliation(s)
- Lorico Ds Lapitan
- Department of Chemical Engineering, Faculty of Engineering, University of Santo Tomas, España Boulevard, Manila, Philippines, Center for Advanced Materials and Technologies-CEZAMAT, Warsaw University of Technology, 02-822, Warsaw, Poland
| | - Bren Mark B Felisilda
- Department of Electrode Processes, Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224, Warsaw, Poland, Department of Chemistry, College of Arts & Sciences, Xavier University-Ateneo de Cagayan, Corrales Street, Cagayan de Oro, Philippines
| | - Cristina E Tiangco
- Research Center for the Natural and Applied Sciences and, Department of Chemical Engineering, Faculty of Engineering, University of Santo Tomas, España Boulevard, Manila, Philippines
| | - Ammu Rosin Jose
- Department of Chemistry, Sacred Heart College (Autonomous), Pandit Karuppan Rd, Thevara, Ernakulam, Kerala, India
| |
Collapse
|
12
|
Wiest A, Kielkowski P. Improved deconvolution of natural products' protein targets using diagnostic ions from chemical proteomics linkers. Beilstein J Org Chem 2024; 20:2323-2341. [PMID: 39290210 PMCID: PMC11406061 DOI: 10.3762/bjoc.20.199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Identification of interactions between proteins and natural products or similar active small molecules is crucial for understanding of their mechanism of action on a molecular level. To search elusive, often labile, and low-abundant conjugates between proteins and active compounds, chemical proteomics introduces a feasible strategy that allows to enrich and detect these conjugates. Recent advances in mass spectrometry techniques and search algorithms provide unprecedented depth of proteome coverage and the possibility to detect desired modified peptides with high sensitivity. The chemical 'linker' connecting an active compound-protein conjugate with a detection tag is the critical component of all chemical proteomic workflows. In this review, we discuss the properties and applications of different chemical proteomics linkers with special focus on their fragmentation releasing diagnostic ions and how these may improve the confidence in identified active compound-peptide conjugates. The application of advanced search options improves the identification rates and may help to identify otherwise difficult to find interactions between active compounds and proteins, which may result from unperturbed conditions, and thus are of high physiological relevance.
Collapse
Affiliation(s)
- Andreas Wiest
- LMU Munich, Department of Chemistry, Butenandtstr. 5-13, 81377 Munich, Germany
| | - Pavel Kielkowski
- LMU Munich, Department of Chemistry, Butenandtstr. 5-13, 81377 Munich, Germany
| |
Collapse
|
13
|
Xiao S, Mu M, Feng C, Pan S, Chen N. The application of bacteria-nanomaterial hybrids in antitumor therapy. J Nanobiotechnology 2024; 22:536. [PMID: 39227831 PMCID: PMC11373302 DOI: 10.1186/s12951-024-02793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
Adverse effects and multidrug resistance remain significant obstacles in conventional cancer therapy. Nanomedicines, with their intrinsic properties such as nano-sized dimensions and tunable surface characteristics, have the potential to mitigate the side effects of traditional cancer treatments. While nanomaterials have been widely applied in cancer treatment, challenges such as low targeting efficiency and poor tumor penetration persist. Recent research has shown that anaerobic bacteria exhibit high selectivity for primary tumors and metastatic cancers, offering good safety and superior tumor penetration capabilities. This suggests that combining nanomaterials with bacteria could complement their respective limitations, opening vast potential applications in cancer therapy. The use of bacteria in combination with nanomaterials for anticancer treatments, including chemotherapy, radiotherapy, and photothermal/photodynamic therapy, has contributed to the rapid development of the field of bacterial oncology treatments. This review explores the mechanisms of bacterial tumor targeting and summarizes strategies for synthesizing bacterial-nanomaterial and their application in cancer therapy. The combination of bacterial-nanomaterial hybrids with modern therapeutic approaches represents a promising avenue for future cancer treatment research, with the potential to improve treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Susu Xiao
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Mu
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chenqian Feng
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shulin Pan
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Nianyong Chen
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Groaz E, Modranka J, Ploschik D, Jabgunde A, Froeyen M, Jang MY, Wagenknecht HA, Herdewijn P. Impact of sulfur substitution on biotin binding affinity to streptavidin. Bioorg Chem 2024; 150:107600. [PMID: 38945086 DOI: 10.1016/j.bioorg.2024.107600] [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: 05/16/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024]
Abstract
In this study, we investigated how the replacement of the tetrahydrothiophene ring of biotin with either an oxolane or (methyl)pyrrolidine moiety may affect its molecular interactions, in an effort to identify alternative affinity ligands suitable for in vitro and in vivo applications in synthetic biology. Initial molecular dynamics (MD) simulations suggested the potential formation of a hydrogen bond between either the oxygen or nitrogen atom of the envisaged tetrahydroheteryl analogues and the Thr90 residue of streptavidin, mirroring the sulfur-centered hydrogen bond detected by the crystallographic analysis of the biotin-streptavidin interaction. Therefore, oxy-, aza-, and N-methylazabiotin were readily synthesized starting from chiral five- or six-carbon sugar precursors. Based on fluorescence-based titration experiments using the corresponding fluorescein conjugates, oxybiotin showed a binding behavior similar to biotin with streptavidin, while both amino analogues displayed lower binding capacities. Notably, azabiotin exhibited a pH-dependent interaction profile, demonstrating enhanced binding under acidic conditions but weaker binding under basic pH, which could be exploited for various purposes.
Collapse
Affiliation(s)
- Elisabetta Groaz
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Jakub Modranka
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Damian Ploschik
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Amit Jabgunde
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Mathy Froeyen
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Mi-Yeon Jang
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Hans-Achim Wagenknecht
- Institute of Organic Chemistry, Karlsruhe Institute of Technology (KIT), Fritz-Haber-Weg 6, 76131 Karlsruhe, Germany
| | - Piet Herdewijn
- Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
15
|
Chen J, Wang B, Dasgupta A, Porte C, Eckardt L, Qi J, Weiler M, Lammers T, Rix A, Shi Y, Kiessling F. Aminolysis-mediated single-step surface functionalization of poly (butyl cyanoacrylate) microbubbles for ultrasound molecular imaging. J Nanobiotechnology 2024; 22:528. [PMID: 39218888 PMCID: PMC11367926 DOI: 10.1186/s12951-024-02806-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Molecular ultrasound imaging with actively targeted microbubbles (MB) proved promising in preclinical studies but its clinical translation is limited. To achieve this, it is essential that the actively targeted MB can be produced with high batch-to-batch reproducibility with a controllable and defined number of binding ligands on the surface. In this regard, poly (n-butyl cyanoacrylate) (PBCA)-based polymeric MB have been used for US molecular imaging, however, ligand coupling was mostly done via hydrolysis and carbodiimide chemistry, which is a multi-step procedure with poor reproducibility and low MB yield. Herein, we developed a single-step coupling procedure resulting in high MB yields with minimal batch-to-batch variation. Actively targeted PBCA-MB were generated using an aminolysis protocol, wherein amine-containing cRGD was added to the MB using lithium methoxide as a catalyst. We confirmed the successful conjugation of cRGD on the MB surface, while preserving their structure and acoustic signal. Compared to the conventional hydrolysis protocol, aminolysis resulted in higher MB yields and better reproducibility of coupling efficiency. Optical imaging revealed that under flow conditions, cRGD- and rhodamine-labelled MB, generated by aminolysis, specifically bind to tumor necrosis factor-alpha (TNF-α) activated endothelial cells in vitro. Furthermore, US molecular imaging demonstrated a markedly higher binding of the cRGD-MB than of control MB in TNF-α activated mouse aortas and 4T1 tumors in mice. Thus, using the aminolysis based conjugation approach, important refinements on the production of cRGD-MB could be achieved that will facilitate the production of clinical-scale formulations with excellent binding and ultrasound imaging performance.
Collapse
Affiliation(s)
- Junlin Chen
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Bi Wang
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Céline Porte
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Lisa Eckardt
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Jinwei Qi
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Marek Weiler
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Yang Shi
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
16
|
Weber TM, Özdüzenciler P, Tamgüney G, Pietruszka J. Utilization of a Branched Late-Stage Clickable Biotinylated Chassis on the Example of a Pittsburgh B Analogue. Org Lett 2024; 26:6771-6775. [PMID: 39051841 PMCID: PMC11320650 DOI: 10.1021/acs.orglett.4c02527] [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: 07/10/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Biotinylation is probably the most frequent and practically useful modification of molecules to facilitate selective and highly affine binding to (strept)avidin for immobilization, enrichment, and purification for further (bio)chemical or (bio)physical investigations. We present a protecting-group-free synthesis of a branched biotin bis-azide that enables dual-payload late-stage functionalization with arbitrary alkynes via click chemistry. Utility of the chassis is briefly showcased on the example of a valuable Pittsburgh B analogue, which binds pathological protein aggregates, commonly found in neurodegenerative diseases.
Collapse
Affiliation(s)
- T. Moritz Weber
- Mathematisch-Naturwissenschaftliche
Fakultät, Institut für Bioorganische Chemie, Heinrich-Heine-Universität Düsseldorf
im Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Pelin Özdüzenciler
- Institut
für Biologische Informationsprozesse 7 (IBI-7: Strukturbiochemie), Forschungszentrum Jülich, 52428 Jülich, Germany
- Mathematisch-Naturwissenschaftliche
Fakultät, Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Gültekin Tamgüney
- Institut
für Biologische Informationsprozesse 7 (IBI-7: Strukturbiochemie), Forschungszentrum Jülich, 52428 Jülich, Germany
- Mathematisch-Naturwissenschaftliche
Fakultät, Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Jörg Pietruszka
- Mathematisch-Naturwissenschaftliche
Fakultät, Institut für Bioorganische Chemie, Heinrich-Heine-Universität Düsseldorf
im Forschungszentrum Jülich, 52428 Jülich, Germany
- Institut
für Bio- und Geowissenschaften 1 (IBG-1: Biotechnologie), Forschungszentrum Jülich, 52428 Jülich, Germany
| |
Collapse
|
17
|
Zeng H, Ning W, Liu X, Luo W, Xia N. Unlocking the potential of bispecific ADCs for targeted cancer therapy. Front Med 2024; 18:597-621. [PMID: 39039315 DOI: 10.1007/s11684-024-1072-8] [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: 09/30/2023] [Accepted: 02/08/2024] [Indexed: 07/24/2024]
Abstract
Antibody-drug conjugates (ADCs) are biologically targeted drugs composed of antibodies and cytotoxic drugs connected by linkers. These innovative compounds enable precise drug delivery to tumor cells, minimizing harm to normal tissues and offering excellent prospects for cancer treatment. However, monoclonal antibody-based ADCs still present challenges, especially in terms of balancing efficacy and safety. Bispecific antibodies are alternatives to monoclonal antibodies and exhibit superior internalization and selectivity, producing ADCs with increased safety and therapeutic efficacy. In this review, we present available evidence and future prospects regarding the use of bispecific ADCs for cancer treatment, including a comprehensive overview of bispecific ADCs that are currently in clinical trials. We offer insights into the future development of bispecific ADCs to provide novel strategies for cancer treatment.
Collapse
Affiliation(s)
- Hongye Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Wenjing Ning
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xue Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China.
| | - Wenxin Luo
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, the Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
18
|
Stewart JM, Li S, Tang AA, Klocke MA, Gobry MV, Fabrini G, Di Michele L, Rothemund PWK, Franco E. Modular RNA motifs for orthogonal phase separated compartments. Nat Commun 2024; 15:6244. [PMID: 39080253 PMCID: PMC11289419 DOI: 10.1038/s41467-024-50003-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 06/20/2024] [Indexed: 08/02/2024] Open
Abstract
Recent discoveries in biology have highlighted the importance of protein and RNA-based condensates as an alternative to classical membrane-bound organelles. Here, we demonstrate the design of pure RNA condensates from nanostructured, star-shaped RNA motifs. We generate condensates using two different RNA nanostar architectures: multi-stranded nanostars whose binding interactions are programmed via linear overhangs, and single-stranded nanostars whose interactions are programmed via kissing loops. Through systematic sequence design, we demonstrate that both architectures can produce orthogonal (distinct and immiscible) condensates, which can be individually tracked via fluorogenic aptamers. We also show that aptamers make it possible to recruit peptides and proteins to the condensates with high specificity. Successful co-transcriptional formation of condensates from single-stranded nanostars suggests that they may be genetically encoded and produced in living cells. We provide a library of orthogonal RNA condensates that can be modularly customized and offer a route toward creating systems of functional artificial organelles for the task of compartmentalizing molecules and biochemical reactions.
Collapse
Affiliation(s)
- Jaimie Marie Stewart
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA
| | - Shiyi Li
- Department of Bioengineering, University of California, Los Angeles, CA, USA
| | - Anli A Tang
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA, USA
| | - Melissa Ann Klocke
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA, USA
| | - Martin Vincent Gobry
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark
| | - Giacomo Fabrini
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
- fabriCELL, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Lorenzo Di Michele
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, UK
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
- fabriCELL, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Paul W K Rothemund
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA.
- Department of Bioengineering, California Institute of Technology, Pasadena, USA.
- Department of Computation & Neural Systems, California Institute of Technology, Pasadena, USA.
| | - Elisa Franco
- Department of Bioengineering, University of California, Los Angeles, CA, USA.
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Zou M, Zhou H, Gu L, Zhang J, Fang L. Therapeutic Target Identification and Drug Discovery Driven by Chemical Proteomics. BIOLOGY 2024; 13:555. [PMID: 39194493 DOI: 10.3390/biology13080555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/07/2024] [Accepted: 07/19/2024] [Indexed: 08/29/2024]
Abstract
Throughout the human lifespan, from conception to the end of life, small molecules have an intrinsic relationship with numerous physiological processes. The investigation into small-molecule targets holds significant implications for pharmacological discovery. The determination of the action sites of small molecules provide clarity into the pharmacodynamics and toxicological mechanisms of small-molecule drugs, assisting in the elucidation of drug off-target effects and resistance mechanisms. Consequently, innovative methods to study small-molecule targets have proliferated in recent years, with chemical proteomics standing out as a vanguard development in chemical biology in the post-genomic age. Chemical proteomics can non-selectively identify unknown targets of compounds within complex biological matrices, with both probe and non-probe modalities enabling effective target identification. This review attempts to summarize methods and illustrative examples of small-molecule target identification via chemical proteomics. It delves deeply into the interactions between small molecules and human biology to provide pivotal directions and strategies for the discovery and comprehension of novel pharmaceuticals, as well as to improve the evaluation of drug safety.
Collapse
Affiliation(s)
- Mingjie Zou
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Haiyuan Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Letian Gu
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Jingzi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| | - Lei Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center, Medical School of Nanjing University, Nanjing 210093, China
| |
Collapse
|
20
|
Xu J, Brown NJS, Seol Y, Neuman KC. Heterogeneous distribution of kinesin-streptavidin complexes revealed by mass photometry. SOFT MATTER 2024; 20:5509-5515. [PMID: 38832814 PMCID: PMC11254546 DOI: 10.1039/d3sm01702h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Kinesin-streptavidin complexes are widely used in microtubule-based active-matter studies. The stoichiometry of the complexes is empirically tuned but experimentally challenging to determine. Here, mass photometry measurements reveal heterogenous distributions of kinesin-streptavidin complexes. Our binding model indicates that heterogeneity arises from both the kinesin-streptavidin mixing ratio and the kinesin-biotinylation efficiency.
Collapse
Affiliation(s)
- Jing Xu
- Department of Physics, University of California, Merced, CA 95343, USA.
| | - Nathaniel J S Brown
- Department of Quantitative and Systems Biology, University of California, Merced, CA 95343, USA
| | - Yeonee Seol
- Laboratory of Single Molecule Biophysics, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Keir C Neuman
- Laboratory of Single Molecule Biophysics, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
21
|
Oppenheimer KG, Hager NA, McAtee CK, Filiztekin E, Shang C, Warnick JA, Bruchez MP, Brodsky JL, Prosser DC, Kwiatkowski AV, O’Donnell AF. Optimization of the fluorogen-activating protein tag for quantitative protein trafficking and colocalization studies in S. cerevisiae. Mol Biol Cell 2024; 35:mr5. [PMID: 38809589 PMCID: PMC11244157 DOI: 10.1091/mbc.e24-04-0174] [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: 04/19/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 05/30/2024] Open
Abstract
Spatial and temporal tracking of fluorescent proteins (FPs) in live cells permits visualization of proteome remodeling in response to extracellular cues. Historically, protein dynamics during trafficking have been visualized using constitutively active FPs fused to proteins of interest. While powerful, such FPs label all cellular pools of a protein, potentially masking the dynamics of select subpopulations. To help study protein subpopulations, bioconjugate tags, including the fluorogen activation proteins (FAPs), were developed. FAPs are comprised of two components: a single-chain antibody (SCA) fused to the protein of interest and a malachite-green (MG) derivative, which fluoresces only when bound to the SCA. Importantly, the MG derivatives can be either cell-permeant or -impermeant, thus permitting isolated detection of SCA-tagged proteins at the cell surface and facilitating quantitative endocytic measures. To expand FAP use in yeast, we optimized the SCA for yeast expression, created FAP-tagging plasmids, and generated FAP-tagged organelle markers. To demonstrate FAP efficacy, we coupled the SCA to the yeast G-protein coupled receptor Ste3. We measured Ste3 endocytic dynamics in response to pheromone and characterized cis- and trans-acting regulators of Ste3. Our work significantly expands FAP technology for varied applications in S. cerevisiae.
Collapse
Affiliation(s)
| | - Natalie A. Hager
- Department of Biological Sciences, University of Pittsburgh, PA 15260
| | - Ceara K. McAtee
- Department of Biological Sciences, University of Pittsburgh, PA 15260
| | - Elif Filiztekin
- Department of Biological Sciences, University of Pittsburgh, PA 15260
| | - Chaowei Shang
- Department of Biological Sciences, University of Pittsburgh, PA 15260
| | | | - Marcel P. Bruchez
- Molecular Biosensor and Imaging Center, Carnegie Mellon University, Pittsburgh, PA 15213
| | | | - Derek C. Prosser
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Adam V. Kwiatkowski
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261
| | | |
Collapse
|
22
|
Sangji MH, Lee SR, Sai H, Weigand S, Palmer LC, Stupp SI. Self-Sorting vs Coassembly in Peptide Amphiphile Supramolecular Nanostructures. ACS NANO 2024; 18:15878-15887. [PMID: 38848478 DOI: 10.1021/acsnano.4c03083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
The functionality of supramolecular nanostructures can be expanded if systems containing multiple components are designed to either self-sort or mix into coassemblies. This is critical to gain the ability to craft self-assembling materials that integrate functions, and our understanding of this process is in its early stages. In this work, we have utilized three different peptide amphiphiles with the capacity to form β-sheets within supramolecular nanostructures and found binary systems that self-sort and others that form coassemblies. This was measured using atomic force microscopy to reveal the nanoscale morphology of assemblies and confocal laser scanning microscopy to determine the distribution of fluorescently labeled monomers. We discovered that PA assemblies with opposite supramolecular chirality self-sorted into chemically distinct nanostructures. In contrast, the PA molecules that formed a mixture of right-handed, left-handed, and flat nanostructures on their own were able to coassemble with the other PA molecules. We attribute this phenomenon to the energy barrier associated with changing the handedness of a β-sheet twist in a coassembly of two different PA molecules. This observation could be useful for designing biomolecular nanostructures with dual bioactivity or interpenetrating networks of PA supramolecular assemblies.
Collapse
Affiliation(s)
- M Hussain Sangji
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Sieun Ruth Lee
- Department of Materials Science and Engineering, Northwestern University, 2220 Campus Drive, Evanston, Illinois 60208, United States
| | - Hiroaki Sai
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E Superior St., Chicago, Illinois 60611, United States
| | - Steven Weigand
- DuPont-Northwestern-Dow Collaborative Access Team Synchrotron Research Center, Northwestern University, Advanced Photon Source/Argonne National Laboratory 432-A004, Argonne, Illinois 60439, United States
| | - Liam C Palmer
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E Superior St., Chicago, Illinois 60611, United States
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Samuel I Stupp
- Department of Biomedical Engineering, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Department of Materials Science and Engineering, Northwestern University, 2220 Campus Drive, Evanston, Illinois 60208, United States
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, 303 E Superior St., Chicago, Illinois 60611, United States
- Department of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Department of Medicine, Northwestern University, 676 N St. Clair Street, Chicago, Illinois 60611, United States
| |
Collapse
|
23
|
Huang G, Li C, Wu R, Xue G, Song Q, Lan L, Xue C, Xu L, Shen Z. Self-assembly of protein-DNA hybrids dedicated to an accelerated and self-primed strand displacement amplification for reinforced serum microRNA probing. Anal Chim Acta 2024; 1308:342667. [PMID: 38740453 DOI: 10.1016/j.aca.2024.342667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND High-efficiency and highly reliable analysis of microRNAs (miRNAs) in bodily fluids highlights its significance to be extensively utilized as candidates for non-invasive "liquid biopsy" approaches. DNA biosensors based on strand displacement amplification (SDA) methods have been successfully designed to detect miRNAs given the efficiently amplified and recycled of the target sequences. However, the unpredictable DNA framework and heavy reliance on free diffusion or random reactant collisions in existing approaches lead to delayed reaction kinetics and inadequate amplification. Thus, it is crucial to create a modular probe with a controlled structure, high local concentration, and ease of synthesis. RESULTS Inspired by the natural spatial-confinement effect based on a well-known streptavidin-biotin interaction, we constructed a protein-DNA hybrid, named protein-scaffolded DNA tetrads (PDT), which consists of four biotinylated Y-shaped DNA (Y-DNA) surrounding a streptavidin protein center via a streptavidin-biotin bridge. The streptavidin-biotin recognition system significantly increased the local concentration and intermolecular distance of the probes to achieve enhanced reaction efficiency and kinetics. The PDT-based assay starts with the target miRNA binding to Y-DNA, which disassembles the Y-DNA structures into three types of hairpin-shaped structures via self-primed strand displacement amplification (SPSDA) and generates remarkable fluorescence signal that is proportional to the miRNA concentration. Results demonstrated that PDT enabled a more efficient detection of miRNA-21 with a sensitivity of 1 fM. Moreover, it was proven reliable for the detection of clinical serum samples, suggesting great potential for advancing the development of rapid and robust signal amplification technologies for early diagnosis. SIGNIFICANCE This simple yet robust system contributes to the early diagnosis of miR-21 with satisfactory sensitivity and specificity, and display a significantly improved nuclease resistance owing to their unique structure. The results suggested that the strategy is expected to provide a promising potential platform for tumor diagnosis, prognosis and therapy.
Collapse
Affiliation(s)
- Guoqiao Huang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, PR China; Department of Laboratory Medicine, Jintang First People's Hospital, West China Hospital Sichuan University Jintang Hospital, Chengdu, Sichuan, 610400, PR China
| | - Chan Li
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Rong Wu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Guohui Xue
- Department of Clinical Laboratory, Jiujiang NO.1 People's Hospital, Jiujiang, Jiangxi, 332000, PR China
| | - Qiufeng Song
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Linwen Lan
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, PR China
| | - Chang Xue
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, PR China.
| | - Liang Xu
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, PR China.
| | - Zhifa Shen
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, PR China.
| |
Collapse
|
24
|
Lim W, Lee S, Koh M, Jo A, Park J. Recent advances in chemical biology tools for protein and RNA profiling of extracellular vesicles. RSC Chem Biol 2024; 5:483-499. [PMID: 38846074 PMCID: PMC11151817 DOI: 10.1039/d3cb00200d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 04/25/2024] [Indexed: 06/09/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized vesicles secreted by cells that contain various cellular components such as proteins, nucleic acids, and lipids from the parent cell. EVs are abundant in body fluids and can serve as circulating biomarkers for a variety of diseases or as a regulator of various biological processes. Considering these characteristics of EVs, analysis of the EV cargo has been spotlighted for disease diagnosis or to understand biological processes in biomedical research. Over the past decade, technologies for rapid and sensitive analysis of EVs in biofluids have evolved, but detection and isolation of targeted EVs in complex body fluids is still challenging due to the unique physical and biological properties of EVs. Recent advances in chemical biology provide new opportunities for efficient profiling of the molecular contents of EVs. A myriad of chemical biology tools have been harnessed to enhance the analytical performance of conventional assays for better understanding of EV biology. In this review, we will discuss the improvements that have been achieved using chemical biology tools.
Collapse
Affiliation(s)
- Woojeong Lim
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
| | - Soyeon Lee
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
| | - Minseob Koh
- Department of Chemistry, Pusan National University Busan 46241 Republic of Korea
| | - Ala Jo
- Center for Nanomedicine, Institute for Basic Science Seoul 03722 Republic of Korea
| | - Jongmin Park
- Department of Chemistry, Kangwon National University Chuncheon 24341 Korea
- Institute for Molecular Science and Fusion Technology, Kangwon National University Chuncheon 24341 Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University Chuncheon 24341 Republic of Korea
| |
Collapse
|
25
|
Alhassan AM, Shirure VS, Luo J, Nguyen BB, Rollins ZA, Shergill BS, Zhu X, Baumgarth N, George SC. A Microfluidic Strategy to Capture Antigen‐Specific High‐Affinity B Cells. ADVANCED NANOBIOMED RESEARCH 2024; 4. [DOI: 10.1002/anbr.202300101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Assessing B cell affinity to pathogen‐specific antigens prior to or following exposure could facilitate the assessment of immune status. Current standard tools to assess antigen‐specific B cell responses focus on equilibrium binding of the secreted antibody in serum. These methods are costly, time‐consuming, and assess antibody affinity under zero force. Recent findings indicate that force may influence BCR‐antigen binding interactions and thus immune status. Herein, a simple laminar flow microfluidic chamber in which the antigen (hemagglutinin of influenza A) is bound to the chamber surface to assess antigen‐specific BCR binding affinity of five hemagglutinin‐specific hybridomas from 65 to 650 pN force range is designed. The results demonstrate that both increasing shear force and bound lifetime can be used to enrich antigen‐specific high‐affinity B cells. The affinity of the membrane‐bound BCR in the flow chamber correlates well with the affinity of the matched antibodies measured in solution. These findings demonstrate that a microfluidic strategy can rapidly assess BCR‐antigen‐binding properties and identify antigen‐specific high‐affinity B cells. This strategy has the potential to both assess functional immune status from peripheral B cells and be a cost‐effective way of identifying individual B cells as antibody sources for a range of clinical applications.
Collapse
Affiliation(s)
- Ahmed M. Alhassan
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| | - Venktesh S. Shirure
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| | - Jean Luo
- Department of Pathology, Microbiology, and Immunology University of California Davis CA 95616 USA
| | - Bryan B. Nguyen
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| | - Zachary A. Rollins
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| | | | - Xiangdong Zhu
- Department of Physics and Astronomy University of California Davis CA 95616 USA
| | - Nicole Baumgarth
- Department of Pathology, Microbiology, and Immunology University of California Davis CA 95616 USA
- Department of Molecular Microbiology and Immunology Bloomberg School of Public Health and Department of Molecular and Comparative Pathobiology School of Medicine Johns Hopkins University Baltimore MD 21205 USA
| | - Steven C. George
- Department of Biomedical Engineering University of California Davis CA 95616 USA
| |
Collapse
|
26
|
Williams TL, Taily IM, Hatton L, Berezin AA, Wu Y, Moliner V, Świderek K, Tsai Y, Luk LYP. Secondary Amine Catalysis in Enzyme Design: Broadening Protein Template Diversity through Genetic Code Expansion. Angew Chem Int Ed Engl 2024; 63:e202403098. [PMID: 38545954 PMCID: PMC11497281 DOI: 10.1002/anie.202403098] [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: 02/13/2024] [Indexed: 04/20/2024]
Abstract
Secondary amines, due to their reactivity, can transform protein templates into catalytically active entities, accelerating the development of artificial enzymes. However, existing methods, predominantly reliant on modified ligands or N-terminal prolines, impose significant limitations on template selection. In this study, genetic code expansion was used to break this boundary, enabling secondary amines to be incorporated into alternative proteins and positions of choice. Pyrrolysine analogues carrying different secondary amines could be incorporated into superfolder green fluorescent protein (sfGFP), multidrug-binding LmrR and nucleotide-binding dihydrofolate reductase (DHFR). Notably, the analogue containing a D-proline moiety demonstrated both proteolytic stability and catalytic activity, conferring LmrR and DHFR with the desired transfer hydrogenation activity. While the LmrR variants were confined to the biomimetic 1-benzyl-1,4-dihydronicotinamide (BNAH) as the hydride source, the optimal DHFR variant favorably used the pro-R hydride from NADPH for stereoselective reactions (e.r. up to 92 : 8), highlighting that a switch of protein template could broaden the nucleophile option for catalysis. Owing to the cofactor compatibility, the DHFR-based secondary amine catalysis could be integrated into an enzymatic recycling scheme. This established method shows substantial potential in enzyme design, applicable from studies on enzyme evolution to the development of new biocatalysts.
Collapse
Affiliation(s)
- Thomas L. Williams
- School of Chemistry and Cardiff Catalysis InstituteCardiff UniversityMain Building, Park PlaceCardiffCF10 3ATUnited Kingdom
| | - Irshad M. Taily
- School of Chemistry and Cardiff Catalysis InstituteCardiff UniversityMain Building, Park PlaceCardiffCF10 3ATUnited Kingdom
| | - Lewis Hatton
- School of Chemistry and Cardiff Catalysis InstituteCardiff UniversityMain Building, Park PlaceCardiffCF10 3ATUnited Kingdom
| | - Andrey A Berezin
- School of Chemistry and Cardiff Catalysis InstituteCardiff UniversityMain Building, Park PlaceCardiffCF10 3ATUnited Kingdom
| | - Yi‐Lin Wu
- School of Chemistry and Cardiff Catalysis InstituteCardiff UniversityMain Building, Park PlaceCardiffCF10 3ATUnited Kingdom
| | - Vicent Moliner
- BioComp Group, Institute of Advanced Materials (INAM)Universitat Jaume I12071CastellóSpain
| | - Katarzyna Świderek
- BioComp Group, Institute of Advanced Materials (INAM)Universitat Jaume I12071CastellóSpain
| | - Yu‐Hsuan Tsai
- Institute of Molecular PhysiologyShenzhen Bay LaboratoryGaoke International Innovation CenterGuangming District518132Shenzhen, GuangdongChina
| | - Louis Y. P. Luk
- School of Chemistry and Cardiff Catalysis InstituteCardiff UniversityMain Building, Park PlaceCardiffCF10 3ATUnited Kingdom
| |
Collapse
|
27
|
Saipul Bahri NSN, Nguyen TT, Matsumoto K, Watanabe M, Morita Y, Septiani EL, Cao KLA, Hirano T, Ogi T. Controlling the Magnetic Responsiveness of Cellulose Nanofiber Particles Embedded with Iron Oxide Nanoparticles. ACS APPLIED BIO MATERIALS 2024; 7:3227-3237. [PMID: 38627897 DOI: 10.1021/acsabm.4c00213] [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] [Indexed: 05/21/2024]
Abstract
2,2,6,6-tetramethylpiperidine-1-oxyl (TEMPO)-oxidized cellulose nanofiber (TOCN) particles, an innovative biobased material derived from wood biomass, have garnered significant interest, particularly in the biomedical field, for their distinctive properties as biocompatible particle adsorbents. However, their microscopic size complicates their separation in liquid media, thereby impeding their application in various domains. In this study, superparamagnetic magnetite nanoparticles (NPs), specifically iron oxide Fe3O4 NPs with an average size of 15 nm, were used to enhance the collection efficiency of TOCN-Fe3O4 composite particles synthesized through spray drying. These composite particles exhibited a remarkable ζ-potential (approximately -50 mV), indicating their high stability in water, as well as impressive magnetization properties (up to 47 emu/g), and rapid magnetic responsiveness within 60 s in water (3 wt % Fe3O4 to TOCN, 1 T magnet). Furthermore, the influence of Fe3O4 NP concentrations on the measurement of the speed of magnetic separation was quantitatively discussed. Additionally, the binding affinity of the synthesized particles for proteins was assessed on a streptavidin-biotin binding system, offering crucial insights into their binding capabilities with specific proteins and underscoring their significant potential as functionalized biomedical materials.
Collapse
Affiliation(s)
- Nur Syakirah Nabilah Saipul Bahri
- Chemical Engineering Program, Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8527, Japan
| | - Tue Tri Nguyen
- Chemical Engineering Program, Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8527, Japan
| | - Kohei Matsumoto
- Life Sciences Headquarters, DKS Co. Ltd., 5 Ogawara, Kisshoin, Minami, Kyoto 601-8391, Japan
| | - Mai Watanabe
- Life Sciences Headquarters, DKS Co. Ltd., 5 Ogawara, Kisshoin, Minami, Kyoto 601-8391, Japan
| | - Yuko Morita
- Life Sciences Headquarters, DKS Co. Ltd., 5 Ogawara, Kisshoin, Minami, Kyoto 601-8391, Japan
| | - Eka Lutfi Septiani
- Chemical Engineering Program, Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8527, Japan
| | - Kiet Le Anh Cao
- Chemical Engineering Program, Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8527, Japan
| | - Tomoyuki Hirano
- Chemical Engineering Program, Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8527, Japan
| | - Takashi Ogi
- Chemical Engineering Program, Department of Advanced Science and Engineering, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8527, Japan
| |
Collapse
|
28
|
Liu K, Wang L, Peng J, Lyu Y, Li Y, Duan D, Zhang W, Wei G, Li T, Niu Y, Zhao Y. Drug-Loaded Bacillus Calmette-Guérin Bacteria for Immuno-Chemo Combo Therapy in Bladder Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310735. [PMID: 38330363 DOI: 10.1002/adma.202310735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/02/2024] [Indexed: 02/10/2024]
Abstract
Intravesical Bacillus Calmette-Guérin (BCG) is a well-established strategy for managing high-risk nonmuscle-invasive bladder cancer (NMIBC); however, over half of patients still experience disease recurrence or progression. Although the combined intravesical instillation of various chemotherapeutic drugs is implemented in clinical trials to enhance the BCG therapy, the outcome is far from satisfying due to severe irritative effects and treatment intolerance at high doses. Therefore, it is adopted the "biotin-streptavidin strategy" to doxorubicin (DOX)-encapsulated nanoparticles within live BCG bacteria (DOX@BCG) to improve treatment outcomes. Adherence of BCG to the bladder epithelium helps precisely target DOX@BCG to the local tumor cells and simultaneously increases intratumoral transport of therapeutic drugs. DOX@BCG effectively inhibits cancer progression and prolongs the survival of rats/mice with orthotopic bladder cancer owing to synergism between BCG-immunotherapy, DOX-chemotherapy, and DOX-induced immunogenic tumor cell death; furthermore, it exhibits improved tolerance and biosafety, and establishes antitumor immunity in the tumor microenvironment. Therefore, the drug-loaded live BCG bacterial delivery system holds considerable potential for clinical translation in the intravesical treatment of bladder cancer.
Collapse
Affiliation(s)
- Kangkang Liu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Lining Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Jing Peng
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yuanji Lyu
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yiming Li
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Dengyi Duan
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Wenyi Zhang
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Guojiang Wei
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Taipeng Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yuanjie Niu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Yang Zhao
- Department of Radiology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| |
Collapse
|
29
|
Zakiyyah SN, Irkham, Einaga Y, Gultom NS, Fauzia RP, Kadja GTM, Gaffar S, Ozsoz M, Hartati YW. Green Synthesis of Ceria Nanoparticles from Cassava Tubers for Electrochemical Aptasensor Detection of SARS-CoV-2 on a Screen-Printed Carbon Electrode. ACS APPLIED BIO MATERIALS 2024; 7:2488-2498. [PMID: 38577953 DOI: 10.1021/acsabm.4c00088] [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] [Indexed: 04/06/2024]
Abstract
Green synthesis approaches for making nanosized ceria using starch from cassava as template molecules to control the particle size are reported. The results of the green synthesis of ceria with an optimum calcination temperature of 800 °C shows a size distribution of each particle of less than 30 nm with an average size of 9.68 nm, while the ratio of Ce3+ to Ce4+ was 25.6%. The green-synthesized nanoceria are applied to increase the sensitivity and attach biomolecules to the electrode surface of the electrochemical aptasensor system for coronavirus disease (COVID-19). The response of the aptasensor to the receptor binding domain of the virus was determined with the potassium ferricyanide redox system. The screen-printed carbon electrode that has been modified with green-synthesized nanoceria shows 1.43 times higher conductivity than the bare electrode, while those modified with commercial ceria increase only 1.18 times. Using an optimized parameter for preparing the aptasensors, the detection and quantification limits were 1.94 and 5.87 ng·mL-1, and the accuracy and precision values were 98.5 and 89.1%. These results show that green-synthesized ceria could be a promising approach for fabricating an electrochemical aptasensor.
Collapse
Affiliation(s)
- Salma Nur Zakiyyah
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University, Jl. Raya Bandung-Sumedang Km 21, Jatinangor, Sumedang, West Java 45363, Indonesia
| | - Irkham
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University, Jl. Raya Bandung-Sumedang Km 21, Jatinangor, Sumedang, West Java 45363, Indonesia
| | - Yasuaki Einaga
- Department of Chemistry, Keio University, 3-14-1 Hiyoshi, Yokohama, 223-8522, Japan
| | - Noto Susanto Gultom
- Department of Physics, Faculty of Mathematics and Natural Sciences, Padjadjaran University, Jl. Raya Bandung-Sumedang Km 21, Jatinangor, Sumedang, West Java 45363, Indonesia
| | - Retna Putri Fauzia
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University, Jl. Raya Bandung-Sumedang Km 21, Jatinangor, Sumedang, West Java 45363, Indonesia
| | - Grandprix Thomreys Marth Kadja
- Division of Inorganic and Physical Chemistry, Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jl. Ganesha no. 10, Bandung 40132, Indonesia
- Research Center for Nanosciences and Nanotechnology, Institut Teknologi Bandung, Jl. Ganesha no. 10, Bandung 40132, Indonesia
| | - Shabarni Gaffar
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University, Jl. Raya Bandung-Sumedang Km 21, Jatinangor, Sumedang, West Java 45363, Indonesia
| | - Mehmet Ozsoz
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University, Jl. Raya Bandung-Sumedang Km 21, Jatinangor, Sumedang, West Java 45363, Indonesia
- Department of Biomedical Engineering, Near East University, Mersin 99138, Turkey
| | - Yeni Wahyuni Hartati
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Padjadjaran University, Jl. Raya Bandung-Sumedang Km 21, Jatinangor, Sumedang, West Java 45363, Indonesia
| |
Collapse
|
30
|
Talapphet N, Huh CS, Kim MM. Development of gold nanocluster complex for the detection of tumor necrosis factor-alpha based on immunoassay. J Immunol Methods 2024; 527:113648. [PMID: 38373541 DOI: 10.1016/j.jim.2024.113648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/13/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Tumor necrosis factor-alpha, TNF-α, a cytokine recognized as a key regulator of inflammatory responses, is primarily produced by activated monocytes and macrophages. Measuring TNF-α levels serves as a valuable indicator for tracking several diseases and pathological states. Gold nanotechnology has been identified as a highly effective catalyst with unique properties for measuring inflammatory cytokines. This study aimed to synthesize gold nanoclusters (AuNCs) and the AuNCs-streptavidin system, along with their characterizations and spherical morphology. The detection of TNF-α antigen with AuNCs was determined, and a new immunoassay-based AuNCs analytical platform was studied. In this study, it was demonstrated that the synthesized AuNCs and AuNCs-streptavidin showed a bright-yellow appearance with absorption peaks at A600 and A610 nm, respectively. The approximately spherical shape was observed by TEM analysis. The AuNCs demonstrated a sensitivity limit for the detection of the TNF-α antigen, with a linear dose-dependent detection range of less than 1.25 ng/mL. The products of the band sizes and band intensities were proportional to the amount of TNF-α in the range of ∼80 kDa, ∼55 kDa, and ∼ 25 kDa in western blot analysis. The TNF-α in cell lysate was successfully detected using an immunoassay after the activation of RAW264.7 cells with lipopolysaccharide (LPS). This assay may serve as a viable alternative for TNF-α detection with high speed, sensitivity, and qualities, ensuring its broad applications.
Collapse
Affiliation(s)
- Natchanok Talapphet
- Department of Applied Chemistry, Dong-Eui University, Busan 47340, Republic of Korea
| | - Chang Soon Huh
- Department of Applied Chemistry, Dong-Eui University, Busan 47340, Republic of Korea
| | - Moon-Moo Kim
- Department of Applied Chemistry, Dong-Eui University, Busan 47340, Republic of Korea.
| |
Collapse
|
31
|
Zhang Y, Zhang Y, Zhou W, He P, Sun X, Li J, Wei H, Yu J. Rapid and sensitive detection of SARS-CoV-2 IgM through luciferase luminescence on an automatic platform. Int J Biol Macromol 2024; 265:130964. [PMID: 38499123 DOI: 10.1016/j.ijbiomac.2024.130964] [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: 01/19/2024] [Revised: 02/27/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
SARS-CoV-2 has brought a global health crisis worldwide. IgM is an early marker in sera after the infections, and the detection of IgM is crucial to assist diagnosis and evaluate the vaccination clinically. Herein, we developed an automated platform to identify IgM against SARS-CoV-2 in sera. Streptavidin-magnetic beads were utilized to bind to a biotinylated anti-IgM antibody, which was employed to capture IgM in sera. RBD fused luciferase hGluc was employed to label the trapped IgM against RBD and the signal of luminescence of hGluc with the substrate of coelenterazine corresponded to the amount of SARS-CoV-2 IgM conjugated to the magnetic beads. An appropriate cut-off value of the designed method was defined by a set of negative samples and positive samples with 100 % sensitivity and 100 % specificity. Through serial dilution of a positive sample, it was found that the method has a better sensitivity than ELISA. The application to determine IgM against SARS-CoV-2 demonstrated a good performance of the method. The developed system can complete the analysis of SARS-CoV-2 IgM within 25 min. Through the substitution of RBD antigen with antigens of other pathogens in this platform, the automated detection of IgM against the corresponding pathogens can be realized.
Collapse
Affiliation(s)
- Yibing Zhang
- Laboratory of Infection and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, PR China
| | - Yun Zhang
- Laboratory of Infection and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, PR China
| | - Wenhao Zhou
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ping He
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Xueni Sun
- Laboratory of Infection and Immunology, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, PR China
| | - Junhua Li
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, PR China
| | - Hongping Wei
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Junping Yu
- CAS Key Laboratory of Special Pathogens and Biosafety, Center for Emerging Infectious Diseases, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| |
Collapse
|
32
|
Liao Z, Jiang J, Wu W, Shi J, Wang Y, Yao Y, Sheng T, Liu F, Liu W, Zhao P, Lv F, Sun J, Li H, Gu Z. Lymph node-biomimetic scaffold boosts CAR-T therapy against solid tumor. Natl Sci Rev 2024; 11:nwae018. [PMID: 38440217 PMCID: PMC10911814 DOI: 10.1093/nsr/nwae018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/10/2023] [Accepted: 01/08/2024] [Indexed: 03/06/2024] Open
Abstract
The limited infiltration and persistence of chimeric antigen receptor (CAR)-T cells is primarily responsible for their treatment deficits in solid tumors. Here, we present a three-dimensional scaffold, inspired by the physiological process of T-cell proliferation in lymph nodes. This scaffold gathers the function of loading, delivery, activation and expansion for CAR-T cells to enhance their therapeutic effects on solid tumors. This porous device is made from poly(lactic-co-glycolic acid) by a microfluidic technique with the modification of T-cell stimulatory signals, including anti-CD3, anti-CD28 antibodies, as well as cytokines. This scaffold fosters a 50-fold CAR-T cell expansion in vitro and a 15-fold cell expansion in vivo. Particularly, it maintains long-lasting expansion of CAR-T cells for up to 30 days in a cervical tumor model and significantly inhibits the tumor growth. This biomimetic delivery strategy provides a versatile platform of cell delivery and activation for CAR-T cells in treating solid tumors.
Collapse
Affiliation(s)
- Ziyan Liao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Jie Jiang
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei Wu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jiaqi Shi
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Yanfang Wang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yuejun Yao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tao Sheng
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Feng Liu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Wei Liu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Feifei Lv
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Sun
- Bone Marrow Transplantation Center of the First Affiliated Hospital and Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Hongjun Li
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Zhen Gu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
33
|
Liu J, Yuan S, Bremmer A, Hu Q. Convergence of Nanotechnology and Bacteriotherapy for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309295. [PMID: 38358998 PMCID: PMC11040386 DOI: 10.1002/advs.202309295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/01/2024] [Indexed: 02/17/2024]
Abstract
Bacteria have distinctive properties that make them ideal for biomedical applications. They can self-propel, sense their surroundings, and be externally detected. Using bacteria as medical therapeutic agents or delivery platforms opens new possibilities for advanced diagnosis and therapies. Nano-drug delivery platforms have numerous advantages over traditional ones, such as high loading capacity, controlled drug release, and adaptable functionalities. Combining bacteria and nanotechnologies to create therapeutic agents or delivery platforms has gained increasing attention in recent years and shows promise for improved diagnosis and treatment of diseases. In this review, design principles of integrating nanoparticles with bacteria, bacteria-derived nano-sized vesicles, and their applications and future in advanced diagnosis and therapeutics are summarized.
Collapse
Affiliation(s)
- Jun Liu
- Pharmaceutical Sciences Division, School of PharmacyUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
- Wisconsin Center for NanoBioSystemsUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
- Carbone Cancer Center, School of Medicine and Public HealthUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
| | - Sichen Yuan
- Pharmaceutical Sciences Division, School of PharmacyUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
- Wisconsin Center for NanoBioSystemsUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
- Carbone Cancer Center, School of Medicine and Public HealthUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
| | - Alexa Bremmer
- Pharmaceutical Sciences Division, School of PharmacyUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of PharmacyUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
- Wisconsin Center for NanoBioSystemsUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
- Carbone Cancer Center, School of Medicine and Public HealthUniversity of Wisconsin, Madison (UW‐Madison)MadisonWI53705USA
| |
Collapse
|
34
|
Lee J, Soares G, Doty C, Park J, Hovey J, Schrader A, Han HS. Versatile Prepolymer Platform for Controlled Tailoring of Quantum Dot Surface Properties. ACS APPLIED MATERIALS & INTERFACES 2024; 16:15202-15214. [PMID: 38470982 PMCID: PMC11070902 DOI: 10.1021/acsami.4c00226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Quantum dots (QDs) hold immense promise for bioimaging, yet technical challenges in surface engineering limit their wider scientific use. We introduce poly(pentafluorophenyl acrylate) (PPFPA) as a user-friendly prepolymer platform for creating precisely controlled multidentate polymeric ligands for QD surface engineering, accessible to researchers without extensive synthetic expertise. PPFPA combines the benefits of both bottom-up and prepolymer approaches, offering minimal susceptibility to hydrolysis and side reactions for controlled chemical composition, along with simple synthetic procedures using commercially available reagents. Live cell imaging experiments highlighted a significant reduction in nonspecific binding when employing PPFPA, owing to its minimal hydrolysis, in contrast to ligands synthesized by using a conventional prepolymer prone to uncontrolled hydrolysis. This observation underscores the distinct advantage of our prepolymer system. Leveraging PPFPA, we synthesized biomolecule-conjugated QDs and performed QD-based immunofluorescence to detect a cytosolic protein. To effectively label cytosolic targets in such a dense and complex environment, probes must exhibit minimal nonspecific binding and be compact. As a result, QD-immunofluorescence has focused primarily on cell surface targets. By creating compact QD-F(ab')2, we sensitively detected alpha-tubulin with a ∼50-fold higher signal-to-noise ratio compared to organic dye-based labeling. PPFPA represents a versatile and accessible platform for tailoring QD surfaces, offering a pathway to realize the full potential of colloidal QDs in various scientific applications.
Collapse
Affiliation(s)
- JuYeon Lee
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- School of Biological and Environmental Studies, Millikin University, 1184 W. Main Street, Decatur, Illinois 62522, United States
| | - Giselle Soares
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Calvin Doty
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Joonhyuck Park
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Jack Hovey
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Alex Schrader
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hee-Sun Han
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
35
|
Zheng Y, Jiang M, Zhu X, Chen Y, Feng L, Zhu H. Metabolic labeling-mediated visualization, capture, and inactivation of Gram-positive bacteria via biotin-streptavidin interactions. Chem Commun (Camb) 2024. [PMID: 38477080 DOI: 10.1039/d4cc00517a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
We introduce a biotinylated D-amino acid probe capable of metabolically incorporating into bacterial PG. Leveraging the robust affinity between biotin and streptavidin, the probe has demonstrated efficacy in imaging, capture, and targeted inactivation of Gram-positive bacteria through synergistic pairings with commercially available streptavidin-modified fluorescent dyes and nanomaterials. The versatility of the probe is underscored by its compatibility with a variety of commercially available streptavidin-modified reagents. This adaptability allows the probe to be applied across diverse scenarios by integrating with these commercial reagents.
Collapse
Affiliation(s)
- Yongfang Zheng
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou 350007, China.
| | - Mingyi Jiang
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou 350007, China.
| | - Xinyu Zhu
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou 350007, China.
| | - Yuyuan Chen
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou 350007, China.
| | - Lisha Feng
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou 350007, China.
| | - Hu Zhu
- Fujian-Taiwan Science and Technology Cooperation Base of Biomedical Materials and Tissue Engineering, Engineering Research Center of Industrial Biocatalysis, Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Polymer Materials, College of Chemistry and Materials Science, Fujian Normal University, 32 Shangsan Road, Fuzhou 350007, China.
| |
Collapse
|
36
|
Dukhno O, Ghosh S, Greiner V, Bou S, Godet J, Muhr V, Buchner M, Hirsch T, Mély Y, Przybilla F. Targeted Single Particle Tracking with Upconverting Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2024; 16:11217-11227. [PMID: 38386424 DOI: 10.1021/acsami.3c17116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Single particle tracking (SPT) is a powerful technique for real-time microscopic visualization of the movement of individual biomolecules within or on the surface of living cells. However, SPT often suffers from the suboptimal performance of the photon-emitting labels used to tag the biomolecules of interest. For example, fluorescent dyes have poor photostability, while quantum dots suffer from blinking that hampers track acquisition and interpretation. Upconverting nanoparticles (UCNPs) have recently emerged as a promising anti-Stokes luminescent label for SPT. In this work, we demonstrated targeted SPT using UCNPs. For this, we synthesized 30 nm diameter doped UCNPs and coated them with amphiphilic polymers decorated with polyethylene glycol chains to make them water-dispersible and minimize their nonspecific interactions with cells. Coated UCNPs highly homogeneous in brightness (as confirmed by a single particle investigation) were functionalized by immunoglobulin E (IgE) using a biotin-streptavidin strategy. Using these IgE-UCNP SPT labels, we tracked high-affinity IgE receptors (FcεRI) on the membrane of living RBL-2H3 mast cells at 37 °C in the presence and absence of antigen and obtained good agreement with the literature. Moreover, we used the FcεRI-IgE receptor-antibody system to directly compare the performance of UCNP-based SPT labels to organic dyes (AlexaFluor647) and quantum dots (QD655). Due to their photostability as well as their backgroundless and continuous luminescence, SPT trajectories obtained with UCNP labels are no longer limited by the photophysics of the label but only by the dynamics of the system and, in particular, the movement of the label out of the field of view and/or focal plane.
Collapse
Affiliation(s)
- Oleksii Dukhno
- Laboratory of Biomaging and Pathologies, UMR 7021 CNRS, University of Strasbourg, Strasbourg 67000, France
| | - Srijayee Ghosh
- Laboratory of Biomaging and Pathologies, UMR 7021 CNRS, University of Strasbourg, Strasbourg 67000, France
| | - Vanille Greiner
- Laboratory of Biomaging and Pathologies, UMR 7021 CNRS, University of Strasbourg, Strasbourg 67000, France
| | - Sophie Bou
- Laboratory of Biomaging and Pathologies, UMR 7021 CNRS, University of Strasbourg, Strasbourg 67000, France
| | - Julien Godet
- Laboratory of Biomaging and Pathologies, UMR 7021 CNRS, University of Strasbourg, Strasbourg 67000, France
- IMAGeS team at ICube, UMR 7357, CNRS, Université de Strasbourg, Strasbourg 67000, France
| | - Verena Muhr
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93040 Regensburg, Germany
| | - Markus Buchner
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93040 Regensburg, Germany
| | - Thomas Hirsch
- Institute of Analytical Chemistry, Chemo- and Biosensors, University of Regensburg, 93040 Regensburg, Germany
| | - Yves Mély
- Laboratory of Biomaging and Pathologies, UMR 7021 CNRS, University of Strasbourg, Strasbourg 67000, France
| | - Frédéric Przybilla
- Laboratory of Biomaging and Pathologies, UMR 7021 CNRS, University of Strasbourg, Strasbourg 67000, France
| |
Collapse
|
37
|
Morrow JP, Mazrad ZAI, Warne NM, Ayton S, Bush AI, Kempe K. Schiff-Base Cross-Linked Poly(2-oxazoline) Micelle Drug Conjugates Possess Antiferroptosis Activity in Numerous In Vitro Cell Models. Biomacromolecules 2024; 25:1068-1083. [PMID: 38178625 DOI: 10.1021/acs.biomac.3c01106] [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] [Indexed: 01/06/2024]
Abstract
A great deal of nanocarriers have been applied to induce ferroptosis in cancer research, yet there are limited examples of nanocarrier formulations to rescue ferroptosis, which can be applied to neurodegeneration, inflammation, liver damage, kidney disease, and more. Here, we present the synthesis, characterization, and in vitro evaluation of pH-responsive, core-cross-linked micelle (CCM) ferrostatin-1 (Fer-1) conjugates with amine, valproic acid, and biotin surface chemistries. Fer-1 release from stable and defined CCM Fer-1 conjugates was quantified, highlighting the sustained release for 24 h. CCM Fer-1 conjugates demonstrated excellent ferroptosis rescue by their antilipid peroxidation activity in a diverse set of cell lines in vitro. Additionally, CCMs showed tunable cell association in SH-SY5Y and translocation across an in vitro blood-brain barrier (BBB) model, highlighting potential brain disease applications. Overall, here, we present a polymeric Fer-1 delivery system to enhance Fer-1 action, which could help in improving Fer-1 action in the treatment of ferroptosis-related diseases.
Collapse
Affiliation(s)
- Joshua P Morrow
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Zihnil A I Mazrad
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Nicole M Warne
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
38
|
Ailuno G, Baldassari S, Balboni A, Pastorino S, Zuccari G, Cortese K, Barbieri F, Drava G, Florio T, Caviglioli G. Development of Biotinylated Liposomes Encapsulating Metformin for Therapeutic Targeting of Inflammation-Based Diseases. Pharmaceutics 2024; 16:235. [PMID: 38399288 PMCID: PMC10893420 DOI: 10.3390/pharmaceutics16020235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/29/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Inflammation is a physiological response to a damaging stimulus but sometimes can be the cause of the onset of neurodegenerative diseases, atherosclerosis, and cancer. These pathologies are characterized by the overexpression of inflammatory markers like endothelial adhesion molecules, such as Vascular Cell Adhesion Molecule-1 (VCAM-1). In the present work, the development of liposomes for therapeutic targeted delivery to inflamed endothelia is described. The idea is to exploit a three-step pretargeting system based on the biotin-avidin high-affinity interaction: the first step involves a previously described biotin derivative bearing a VCAM-1 binding peptide; in the second step, the avidin derivative NeutrAvidinTM, which strongly binds to the biotin moiety, is injected; the final step is the administration of biotinylated liposomes that would bind to NeutravidinTM immobilized onto VCAM-1 overexpressing endothelium. Stealth biotinylated liposomes, prepared via the thin film hydration method followed by extrusion and purification via size exclusion chromatography, have been thoroughly characterized for their chemico-physical and morphological features and loaded with metformin hydrochloride, a potential anti-inflammatory agent. The three-step system, tested in vitro on different cell lines via confocal microscopy, FACS analysis and metformin uptake, has proved its suitability for therapeutic applications.
Collapse
Affiliation(s)
- Giorgia Ailuno
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Sara Baldassari
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Alice Balboni
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Sara Pastorino
- Territorial Pharmacy of Azienda Sociosanitaria Ligure 2, Via Carlo Collodi 13, 17100 Savona, Italy;
| | - Guendalina Zuccari
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Katia Cortese
- Department of Experimental Medicine, University of Genoa, Via Antonio de Toni 14, 16132 Genova, Italy;
| | - Federica Barbieri
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV 2, 16132 Genova, Italy; (F.B.); (T.F.)
| | - Giuliana Drava
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| | - Tullio Florio
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV 2, 16132 Genova, Italy; (F.B.); (T.F.)
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| | - Gabriele Caviglioli
- Department of Pharmacy, University of Genoa, Viale Cembrano 4, 16148 Genova, Italy; (S.B.); (A.B.); (G.Z.); (G.D.); (G.C.)
| |
Collapse
|
39
|
Akkaya M, Al Souz J, Williams D, Kamdar R, Kamenyeva O, Kabat J, Shevach E, Akkaya B. Illuminating T cell-dendritic cell interactions in vivo by FlAsHing antigens. eLife 2024; 12:RP91809. [PMID: 38236633 PMCID: PMC10945603 DOI: 10.7554/elife.91809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024] Open
Abstract
Delineating the complex network of interactions between antigen-specific T cells and antigen presenting cells (APCs) is crucial for effective precision therapies against cancer, chronic infections, and autoimmunity. However, the existing arsenal for examining antigen-specific T cell interactions is restricted to a select few antigen-T cell receptor pairs, with limited in situ utility. This lack of versatility is largely due to the disruptive effects of reagents on the immune synapse, which hinder real-time monitoring of antigen-specific interactions. To address this limitation, we have developed a novel and versatile immune monitoring strategy by adding a short cysteine-rich tag to antigenic peptides that emits fluorescence upon binding to thiol-reactive biarsenical hairpin compounds. Our findings demonstrate the specificity and durability of the novel antigen-targeting probes during dynamic immune monitoring in vitro and in vivo. This strategy opens new avenues for biological validation of T-cell receptors with newly identified epitopes by revealing the behavior of previously unrecognized antigen-receptor pairs, expanding our understanding of T cell responses.
Collapse
Affiliation(s)
- Munir Akkaya
- Department of Internal Medicine, Division of Rheumatology and Immunology, The College of Medicine, The Ohio State UniversityColumbusUnited States
- Microbial Infection and Immunity, The Ohio State University Wexner Medical CenterColumbusUnited States
- Pelotonia Institute for Immuno-Oncology, The Ohio State UniversityColumbusUnited States
| | - Jafar Al Souz
- National Institute of Allergy and Infectious DiseasesBethesdaUnited States
| | - Daniel Williams
- National Institute of Allergy and Infectious DiseasesBethesdaUnited States
| | - Rahul Kamdar
- National Institute of Allergy and Infectious DiseasesBethesdaUnited States
| | - Olena Kamenyeva
- National Institute of Allergy and Infectious DiseasesBethesdaUnited States
| | - Juraj Kabat
- National Institute of Allergy and Infectious DiseasesBethesdaUnited States
| | - Ethan Shevach
- National Institute of Allergy and Infectious DiseasesBethesdaUnited States
| | - Billur Akkaya
- Pelotonia Institute for Immuno-Oncology, The Ohio State UniversityColumbusUnited States
- Department of Neurology, The Ohio State University Wexner Medical CenterColumbusUnited States
| |
Collapse
|
40
|
Yang B, Picchetti P, Wang Y, Wang W, Seeger C, Bozov K, Malik S, Mallach D, Schäfer AH, Ibrahim M, Hirtz M, Powell AK. Patterned immobilization of polyoxometalate-loaded mesoporous silica particles via amine-ene Michael additions on alkene functionalized surfaces. Sci Rep 2024; 14:1249. [PMID: 38218940 PMCID: PMC10787769 DOI: 10.1038/s41598-023-50846-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/27/2023] [Indexed: 01/15/2024] Open
Abstract
Polyoxometalates (POM) are anionic oxoclusters of early transition metals that are of great interest for a variety of applications, including the development of sensors and catalysts. A crucial step in the use of POM in functional materials is the production of composites that can be further processed into complex materials, e.g. by printing on different substrates. In this work, we present an immobilization approach for POMs that involves two key processes: first, the stable encapsulation of POMs in the pores of mesoporous silica nanoparticles (MSPs) and, second, the formation of microstructured arrays with these POM-loaded nanoparticles. Specifically, we have developed a strategy that leads to water-stable, POM-loaded mesoporous silica that can be covalently linked to alkene-bearing surfaces by amine-Michael addition and patterned into microarrays by scanning probe lithography (SPL). The immobilization strategy presented facilitates the printing of hybrid POM-loaded nanomaterials onto different surfaces and provides a versatile method for the fabrication of POM-based composites. Importantly, POM-loaded MSPs are useful in applications such as microfluidic systems and sensors that require frequent washing. Overall, this method is a promising way to produce surface-printed POM arrays that can be used for a wide range of applications.
Collapse
Affiliation(s)
- Bingquan Yang
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
- Karlsruhe Nano Micro Facility (KNMFi), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
| | - Pierre Picchetti
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
| | - Yangxin Wang
- Institute of Biological and Chemical Systems - Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
- College of Materials Science and Engineering, Nanjing Tech University, Puzhu Road(S) 30, 211816, Nanjing, People's Republic of China
| | - Wenjing Wang
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
- Karlsruhe Nano Micro Facility (KNMFi), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
| | - Christoph Seeger
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
| | - Kliment Bozov
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
| | - Sharali Malik
- Institute for Quantum Materials and Technologies (IQMT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
| | - Dennis Mallach
- nanoAnalytics GmbH, Heisenbergstraße 11, 48149, Münster, Germany
| | | | - Masooma Ibrahim
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
| | - Michael Hirtz
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany.
- Karlsruhe Nano Micro Facility (KNMFi), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany.
| | - Annie K Powell
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
- Institute of Inorganic Chemistry (AOC), Karlsruhe Institute of Technology (KIT), Engesserstraße 15, 76131, Karlsruhe, Germany
- Institute for Quantum Materials and Technologies (IQMT), Karlsruhe Institute of Technology (KIT), 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
41
|
Zhang M, Luo M, Chen G, Guo H, Zhao J. Study on the properties of a dual-system-based protein scaffold for orthogonal self-assembly. Int J Biol Macromol 2024; 256:127946. [PMID: 37977451 DOI: 10.1016/j.ijbiomac.2023.127946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/06/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
Protein scaffolds possessing the ability to efficiently organize enzymes to improve the catalytic performance, enzyme stability and provide an optimal micro-environment for biocatalysis. Here, SpyCatcher fused to the C-terminus of Treptavidin (a variant of streptavidin) to construct a chimeric tetramers protein scaffold (Tr-SC) with dual orthogonal conjugation moieties. The results showed that the expressed Tr-SC scaffold was an active tetramer with good stability under 80 °C and pH 6.5-8.5, which could bind 4 SpyTag-mCherry and 4 Biotin-EGFP. Tr-SC scaffold can bind 1-4 ligands alone under different conditions. The order in which protein scaffolds bind to proteins has little effect on the final complex structure. It is more difficult for SpyTag-mCherry than Biotin-EGFP to bind to Tr-SC, so incomplete conjugates of a hexameric complex composed of 2 SpyTag-mCherry and 4 Biotin-EGFP form when the molar ratio of scaffold and two ligands is 1:4:4. Therefore, it was suggest that the Tr-SC can first bind to excess SpyTag-protein and mixed with Biotin-protein to promote the formation of higher multimers. The results can be important reference for more extensive use of Tr-SC to construct heterologous protein polymers and assembly of heterologous enzyme molecular machine in vitro to carry on efficient cascade reaction in the future.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Bioengineering and Biotechnology, Huaqiao University, Jimei Ave. 668, Xiamen 361021, China
| | - Mianxing Luo
- Department of Bioengineering and Biotechnology, Huaqiao University, Jimei Ave. 668, Xiamen 361021, China
| | - Guo Chen
- Department of Bioengineering and Biotechnology, Huaqiao University, Jimei Ave. 668, Xiamen 361021, China.
| | - Hongwei Guo
- Department of Bioengineering and Biotechnology, Huaqiao University, Jimei Ave. 668, Xiamen 361021, China
| | - Jun Zhao
- Department of Bioengineering and Biotechnology, Huaqiao University, Jimei Ave. 668, Xiamen 361021, China
| |
Collapse
|
42
|
Shi X, Fan W, Mehrpouyan M, Chen Y, D'Cruz LM, Widmann SJ, Tyznik AJ. Flow cytometry analysis of protein expression using antibody-derived tags followed by CITE-Seq. Cytometry A 2024; 105:62-73. [PMID: 37772953 DOI: 10.1002/cyto.a.24792] [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: 02/19/2023] [Revised: 05/13/2023] [Accepted: 09/04/2023] [Indexed: 09/30/2023]
Abstract
Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-Seq) is a single-cell phenotyping method that uses antibody-derived tags (ADTs) to quantitatively detect cell surface protein expression and generate transcriptomic data at the single-cell level. Despite the increased popularity of this technique to study cellular heterogeneity and dynamics, detailed methods on how to choose ADT markers and ensuring reagent performance in biological relevant systems prior to sequencing is not available. Here we describe a novel and easy-to-use multiplex flow proxy assay in which multiple protein markers can be measured simultaneously using a combination of ADT reagents and dye-oligo conjugates by flow cytometry. Using dye-oligo conjugates with sequences complementary to the ADT reagents, we can achieve specific binding and evaluate protein marker expression in a multiplex way. This quality control assay is useful for guiding ADT marker choice and confirming protein expression prior to sequencing. Importantly, the labeled cells can be directly isolated based on the specific fluorescence from dye-oligo conjugates using a flow cytometry cell sorter and processed for downstream single-cell multiomics. Using this streamlined workflow, we sorted natural killer cells and T cells efficiently using only ADT and dye-oligo reagents, avoiding the possibility of decreased marker resolution from co-staining cells with ADT and fluorescent antibodies. This novel workflow provides a viable option for improving ADT marker choice and cell sorting efficiency, allowing subsequent CITE-Seq.
Collapse
Affiliation(s)
- Xiaoshan Shi
- Applied Research & Technology, Medical Scientific Affairs, BD Biosciences, San Jose, California, USA
| | - Wei Fan
- Chemistry Development, BD Biosciences, San Jose, California, USA
| | - Majid Mehrpouyan
- Chemistry Development, BD Biosciences, San Jose, California, USA
| | - Yu Chen
- Chemistry Development, BD Biosciences, San Diego, California, USA
| | - Louise M D'Cruz
- Applied Research & Technology, Medical Scientific Affairs, BD Biosciences, San Diego, California, USA
| | - Stephanie J Widmann
- Applied Research & Technology, Medical Scientific Affairs, BD Biosciences, San Diego, California, USA
| | - Aaron J Tyznik
- Applied Research & Technology, Medical Scientific Affairs, BD Biosciences, San Diego, California, USA
| |
Collapse
|
43
|
Wang S, Zhou Y, Li Z. A microfluidic cover converts a standard 96-well plate into a mass-transport-controlled immunoassay system. BIOMICROFLUIDICS 2024; 18:014102. [PMID: 38249129 PMCID: PMC10798817 DOI: 10.1063/5.0183651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/01/2024] [Indexed: 01/23/2024]
Abstract
96-well microtiter plates, widely used in immunoassays, face challenges such as prolonged assay time and limited sensitivity due to the lack of analyte transport control. Orbital shakers, commonly employed to facilitate mass transport, offer limited improvements and can introduce assay inconsistencies. While microfluidic devices offer performance enhancements, their complexity and incompatibility with existing platforms limit their wide adoption. This study introduces a novel microfluidic 96-well cover designed to convert a standard 96-well plate to a mass-transport-controlled surface bioreactor. The cover employs microfluidic methods to enhance the diffusion flux of analytes toward the receptors immobilized on the well bottom. Both simulation and experimental results demonstrated that the cover significantly enhances the capture rate of analyte molecules, resulting in increased signal strength for various detection methods and a lower detection limit. The cover serves as an effective add-on to standard 96-well plates, offering enhanced assay performance without requiring modifications to existing infrastructure or reagents. This innovation holds promise for improving the efficiency and reliability of microtiter plate based immunoassays.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Biomedical Engineering, The George Washington University, District of Columbia, 20052, USA
| | - You Zhou
- Department of Electrical and Computer Engineering, The George Washington University, District of Columbia, 20052, USA
| | - Zhenyu Li
- Department of Biomedical Engineering, The George Washington University, District of Columbia, 20052, USA
| |
Collapse
|
44
|
Kang M, Wang Z, Ge X. One-step production of fully biotinylated and glycosylated human Fc gamma receptors. Biotechnol Prog 2024; 40:e3392. [PMID: 37734055 PMCID: PMC10922510 DOI: 10.1002/btpr.3392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023]
Abstract
Initiating and regulating humoral immunity, Fc gamma receptors (FcγRs) have been identified both as therapeutics and as drug targets, and thus production of biologically active FcγRs is highly demanded for biopharmaceutical development. Focusing on low-affinity FcγRs IIA (131H/R allotypes), IIB, and IIIA (176F/V), this study used human 293-F cells to achieve correct post-translational modifications (PTMs) including biotinylation, N-glycosylation, and disulfides. Approaches involving co-expression of FcγR-AviTag and Escherichia coli biotin ligase BirA, endoplasmic reticulum retention, stable and transient transfections, and optimization of transgene ratio were investigated. Protein electrophoresis under reducing and non-reducing conditions, enzymatic deglycosylation, streptavidin pull-down assays, and binding kinetic analysis collectively indicated that the produced FcγR ectodomains were fully biotinylated, N-glycosylated, had formed disulfide bond, and exhibited expected binding affinities toward IgG1 trastuzumab and its Fc mutants. A clear trade-off between production yield and PTM quality was also observed. Achieving multiple types of PTMs completely by one-step cell culture should have applications for the production of a variety of complex proteins of biomedical importance.
Collapse
Affiliation(s)
- Minhyo Kang
- Department of Chemical and Environmental Engineering, University of California Riverside, CA, USA
- Present address: Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC Canada
| | - Zening Wang
- Department of Chemical and Environmental Engineering, University of California Riverside, CA, USA
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, USA
| | - Xin Ge
- Department of Chemical and Environmental Engineering, University of California Riverside, CA, USA
- Institute of Molecular Medicine, University of Texas Health Science Center at Houston, TX, USA
| |
Collapse
|
45
|
Akkaya M, Al Souz J, Williams D, Kamdar R, Kamenyeva O, Kabat J, Shevach EM, Akkaya B. Illuminating T cell-dendritic cell interactions in vivo by FlAsHing antigens. RESEARCH SQUARE 2023:rs.3.rs-3193191. [PMID: 37546912 PMCID: PMC10402196 DOI: 10.21203/rs.3.rs-3193191/v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Delineating the complex network of interactions between antigen-specific T cells and antigen presenting cells (APCs) is crucial for effective precision therapies against cancer, chronic infections, and autoimmunity. However, the existing arsenal for examining antigen-specific T cell interactions is restricted to a select few antigen-T cell receptor pairs, with limited in situ utility. This lack of versatility is largely due to the disruptive effects of reagents on the immune synapse, which hinder real-time monitoring of antigen-specific interactions. To address this limitation, we have developed a novel and versatile immune monitoring strategy by adding a short cysteine-rich tag to antigenic peptides that emits fluorescence upon binding to thiol-reactive biarsenical hairpin compounds. Our findings demonstrate the specificity and durability of the novel antigen-targeting probes during dynamic immune monitoring in vitro and in vivo. This strategy opens new avenues for biological validation of T-cell receptors with newly identified epitopes by revealing the behavior of previously unrecognized antigen-receptor pairs, expanding our understanding of T cell responses.
Collapse
Affiliation(s)
- Munir Akkaya
- Department of Internal Medicine, Division of Rheumatology and Immunology, The College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jafar Al Souz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Williams
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rahul Kamdar
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olena Kamenyeva
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ethan M. Shevach
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Billur Akkaya
- Department of Neurology, The College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
46
|
Akkaya M, Al Souz J, Williams D, Kamdar R, Kamenyeva O, Kabat J, Shevach EM, Akkaya B. Illuminating T cell-dendritic cell interactions in vivo by FlAsHing antigens. RESEARCH SQUARE 2023:rs.3.rs-3193191. [PMID: 37546912 PMCID: PMC10402196 DOI: 10.21203/rs.3.rs-3193191/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Delineating the complex network of interactions between antigen-specific T cells and antigen presenting cells (APCs) is crucial for effective precision therapies against cancer, chronic infections, and autoimmunity. However, the existing arsenal for examining antigen-specific T cell interactions is restricted to a select few antigen-T cell receptor pairs, with limited in situ utility. This lack of versatility is largely due to the disruptive effects of reagents on the immune synapse, which hinder real-time monitoring of antigen-specific interactions. To address this limitation, we have developed a novel and versatile immune monitoring strategy by adding a short cysteine-rich tag to antigenic peptides that emits fluorescence upon binding to thiol-reactive biarsenical hairpin compounds. Our findings demonstrate the specificity and durability of the novel antigen-targeting probes during dynamic immune monitoring in vitro and in vivo. This strategy opens new avenues for biological validation of T-cell receptors with newly identified epitopes by revealing the behavior of previously unrecognized antigen-receptor pairs, expanding our understanding of T cell responses.
Collapse
Affiliation(s)
- Munir Akkaya
- Department of Internal Medicine, Division of Rheumatology and Immunology, The College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jafar Al Souz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Williams
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rahul Kamdar
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olena Kamenyeva
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ethan M. Shevach
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Billur Akkaya
- Department of Neurology, The College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
47
|
Kalita N, Gogoi S, Minteer SD, Goswami P. Advances in Bioelectrode Design for Developing Electrochemical Biosensors. ACS MEASUREMENT SCIENCE AU 2023; 3:404-433. [PMID: 38145027 PMCID: PMC10740130 DOI: 10.1021/acsmeasuresciau.3c00034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 12/26/2023]
Abstract
The critical performance factors such as selectivity, sensitivity, operational and storage stability, and response time of electrochemical biosensors are governed mainly by the function of their key component, the bioelectrode. Suitable design and fabrication strategies of the bioelectrode interface are essential for realizing the requisite performance of the biosensors for their practical utility. A multifaceted attempt to achieve this goal is visible from the vast literature exploring effective strategies for preparing, immobilizing, and stabilizing biorecognition elements on the electrode surface and efficient transduction of biochemical signals into electrical ones (i.e., current, voltage, and impedance) through the bioelectrode interface with the aid of advanced materials and techniques. The commercial success of biosensors in modern society is also increasingly influenced by their size (and hence portability), multiplexing capability, and coupling in the interface of the wireless communication technology, which facilitates quick data transfer and linked decision-making processes in real-time in different areas such as healthcare, agriculture, food, and environmental applications. Therefore, fabrication of the bioelectrode involves careful selection and control of several parameters, including biorecognition elements, electrode materials, shape and size of the electrode, detection principles, and various fabrication strategies, including microscale and printing technologies. This review discusses recent trends in bioelectrode designs and fabrications for developing electrochemical biosensors. The discussions have been delineated into the types of biorecognition elements and their immobilization strategies, signal transduction approaches, commonly used advanced materials for electrode fabrication and techniques for fabricating the bioelectrodes, and device integration with modern electronic communication technology for developing electrochemical biosensors of commercial interest.
Collapse
Affiliation(s)
- Nabajyoti Kalita
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sudarshan Gogoi
- Department
of Chemistry, Sadiya College, Chapakhowa, Assam 786157, India
| | - Shelley D. Minteer
- Department
of Chemistry, University of Utah, 315 S 1400 E, Salt Lake City, Utah 84112, United States
- Kummer
Institute Center for Resource Sustainability, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Pranab Goswami
- Department
of Biosciences and Bioengineering, Indian
Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
48
|
Cheng T, Kosgei BK, Soko GF, Meena SS, Li T, Cao Q, Zhao Z, Cheng SKS, Liu Q, Wang F, Zhu G, Han RPS. Using Functionalized Liposomes to Harvest Extracellular Vesicles of Similar Characteristics in Dermal Interstitial Fluid. Anal Chem 2023; 95:17968-17973. [PMID: 38032052 DOI: 10.1021/acs.analchem.3c04306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Extracellular vesicles (EVs) are used by living cells for the purpose of biological information trafficking from parental-to-recipient cells and vice versa. This back-and-forth communication is enabled by two distinct kinds of biomolecules that constitute the cargo of an EV: proteins and nucleic acids. The proteomic-cum-genetic information is mediated by the physiological state of a cell (healthy or otherwise) as much as modulated by the biogenesis pathway of the EV. Therefore, in mirroring the huge diversities of human communications, the proteins and nucleic acids involved in cell communications possess seemingly near limitless diversities, and it is this characteristic that makes EVs so highly heterogeneous. Currently, there is no simple and reliable tool for the selective capture of heterogeneous EVs and the delivery of their undamaged cargo for research in extracellular protein mapping and spatial proteomics studies. Our work is a preliminary attempt to address this issue. We demonstrated our approach by using antibody functionalized liposomes to capture EVs from tumor and healthy cell-lines. To characterize their performance, we presented fluorescence and nanoparticle tracking analysis (NTA) results, TEM images, and Western blotting analysis for EV proteins. We also extracted dermal interstitial fluid (ISF) from healthy individuals and used our functionalized synthetic vesicle (FSV) method to capture EVs from their proteins. We constructed three proteomic sets [EV vs ISF, (FSV+EV) vs ISF, and (FSV+EV) vs EV] from the EV proteins and the free proteins harvested from ISF and compared their differentially expressed proteins (DEPs). The performance of our proposed method is assessed via an analysis of 1095 proteins, together with volcano plots, heatmap, GO annotation, and enriched KEGG pathways and organelle localization results of 213 DEPs.
Collapse
Affiliation(s)
- Tingjun Cheng
- Jiangzhong Cancer Research Center & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Benson K Kosgei
- Jiangzhong Cancer Research Center & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Geofrey F Soko
- Jiangzhong Cancer Research Center & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
- Ocean Road Cancer Institute, P.O. Box 3592, Dar es Salaam, Tanzania
| | - Stephene S Meena
- Jiangzhong Cancer Research Center & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
- Ocean Road Cancer Institute, P.O. Box 3592, Dar es Salaam, Tanzania
| | - Tong Li
- Jiangzhong Cancer Research Center & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Qianan Cao
- Jiangzhong Cancer Research Center & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Zhe Zhao
- Jiangzhong Cancer Research Center & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Samuel K S Cheng
- School of Engineering, Texas A&M University─Corpus Christi, Corpus Christi, Texas 78412, United States
| | - Qingjun Liu
- Biosensor National Special Laboratory & Key Laboratory for Biomedical Engineering of the Ministry of Education, Dept. of Biomedical Engineering, Zhejiang University, Hangzhou 310027, China
| | - Fang Wang
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Genhua Zhu
- Jiangzhong Cancer Research Center, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| | - Ray P S Han
- Jiangzhong Cancer Research Center & Jiangxi Engineering Research Center for Translational Cancer Technology, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi 330004, China
| |
Collapse
|
49
|
Murillo A, Holgado M, Laguna M. Reports on the sensitivity enhancement in interferometric based biosensors by biotin-streptavidin system. Heliyon 2023; 9:e23123. [PMID: 38149195 PMCID: PMC10750048 DOI: 10.1016/j.heliyon.2023.e23123] [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: 09/08/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/28/2023] Open
Abstract
Antibody biotinylation is a process of attaching biotin molecules to antibodies by chemically modifying specific functional groups on the antibodies without altering their antigen recognition specificity. Biotin, a small vitamin, forms a strong and specific interaction with the protein streptavidin, resulting in a stable biotin-streptavidin (biotin-STV) complex. This biotin-STV interaction is widely exploited in various biotechnological applications, including biosensors. Biosensors are analytical devices that employ biological recognition elements, such as antibodies, enzymes, or nucleic acids, to detect and quantify target analytes in a sample. Antibodies are commonly used as recognition elements in biosensors due to their high specificity and affinity. In this study, the antibody anti-Bovine Serum Albumin (αBSA) has been biotinylated at different antibody:biotin ratios, and the stability of this labeling over time has been investigated. Furthermore, the sensitivity of the biosensor for detecting the Bovine Serum Albumin (BSA) protein has been compared using the biotinylated antibody and the non-biotinylated form, showing a four-fold improvement in detection. This system was also compared with the Enzyme-Linked ImmunoSorbent Assay (ELISA) technique. The advantages of using biotinylated antibodies in biosensors include increased stability and reproducibility of the biorecognition layer, as well as flexibility in sensor design, as different biotinylated antibodies can be utilized for diverse target analytes without altering the sensor's architecture.
Collapse
Affiliation(s)
- A.M.M. Murillo
- Group of Optics, Photonics, and Biophotonics, Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Parque Científico y Tecnológico de La UPM, Campus de Montegancedo, Pozuelo de Alarcón, 28223, Madrid, Spain
- Group of Organ and Tissue on-a-Chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, IdISSC, C/Profesor Martín Lagos s/n, 4a Planta Sur, 28040, Madrid, Spain
| | - M. Holgado
- Group of Optics, Photonics, and Biophotonics, Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Parque Científico y Tecnológico de La UPM, Campus de Montegancedo, Pozuelo de Alarcón, 28223, Madrid, Spain
- Group of Organ and Tissue on-a-Chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, IdISSC, C/Profesor Martín Lagos s/n, 4a Planta Sur, 28040, Madrid, Spain
- Department of Applied Physics and Materials Engineering, Escuela Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, C/José Gutiérrez Abascal, 2, 28006, Madrid, Spain
| | - M. Laguna
- Group of Optics, Photonics, and Biophotonics, Center for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Parque Científico y Tecnológico de La UPM, Campus de Montegancedo, Pozuelo de Alarcón, 28223, Madrid, Spain
- Group of Organ and Tissue on-a-Chip and In-Vitro Detection, Health Research Institute of the Hospital Clínico San Carlos, IdISSC, C/Profesor Martín Lagos s/n, 4a Planta Sur, 28040, Madrid, Spain
- Department of Applied Physics and Materials Engineering, Escuela Técnica Superior de Ingenieros Industriales, Universidad Politécnica de Madrid, C/José Gutiérrez Abascal, 2, 28006, Madrid, Spain
| |
Collapse
|
50
|
Reverte-López M, Gavrilovic S, Merino-Salomón A, Eto H, Yagüe Relimpio A, Rivas G, Schwille P. Protein-Based Patterning to Spatially Functionalize Biomimetic Membranes. SMALL METHODS 2023; 7:e2300173. [PMID: 37350500 DOI: 10.1002/smtd.202300173] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/08/2023] [Indexed: 06/24/2023]
Abstract
The bottom-up reconstitution of proteins for their modular engineering into synthetic cellular systems can reveal hidden protein functions in vitro. This is particularly evident for the bacterial Min proteins, a paradigm for self-organizing reaction-diffusion systems that displays an unexpected functionality of potential interest for bioengineering: the directional active transport of any diffusible cargo molecule on membranes. Here, the MinDE protein system is reported as a versatile surface patterning tool for the rational design of synthetically assembled 3D systems. Employing two-photon lithography, microswimmer-like structures coated with tailored lipid bilayers are fabricated and demonstrate that Min proteins can uniformly pattern bioactive molecules on their surface. Moreover, it is shown that the MinDE system can form stationary patterns inside lipid vesicles, which allow the targeting and distinctive clustering of higher-order protein structures on their inner leaflet. Given their facile use and robust function, Min proteins thus constitute a valuable molecular toolkit for spatially patterned functionalization of artificial biosystems like cell mimics and microcarriers.
Collapse
Affiliation(s)
- María Reverte-López
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Svetozar Gavrilovic
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Adrián Merino-Salomón
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Hiromune Eto
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, 3584 CT, The Netherlands
| | - Ana Yagüe Relimpio
- Department of Cellular Biophysics, Max Planck for Medical Research, 69120, Heidelberg, Germany
| | - Germán Rivas
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, 28040, Spain
| | - Petra Schwille
- Department of Cellular and Molecular Biophysics, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| |
Collapse
|