1
|
Zhang X, Dong Y, Wang Y, Zhang Z, Zhang X, Zhu JJ, Tian Y, Min Q. Quality Control of Mass-Encoded Nanodevices by Compartmented DNA Origami Frames for Precision Information Coding and Logic Mapping. Angew Chem Int Ed Engl 2024; 63:e202313446. [PMID: 38038595 DOI: 10.1002/anie.202313446] [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: 09/10/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/02/2023]
Abstract
Encoded nanostructures afford an ideal platform carrying multi-channel signal components for multiplexed assay and information security. However, with the demand on exclusivity and reproducibility of coding signals, precise control on the structure and composition of nanomaterials featuring fully distinguishable signals remains challenging. By using the multiplexing capability of mass spectrometry (MS) and spatial addressability of DNA origami nanostructures, we herein propose a quality control methodology for constructing mass-encoded nanodevices (namely MNTs-TDOFs) in the scaffold of compartmented tetrahedral DNA origami frames (TDOFs), in which the arrangement and stoichiometry of four types of mass nanotags (MNTs) can be finely regulated and customized to generate characteristic MS patterns. The programmability of combinatorial MNTs and orthogonality of individual compartments allows further evolution of MNTs-TDOFs to static tagging agents and dynamic nanoprobes for labeling and sensing of multiple targets. More importantly, structure control at single TDOF level ensures the constancy of prescribed MS outputs, by which a high-capacity coding system was established for secure information encryption and decryption. In addition to the multiplexed outputs in parallel, the nanodevices could also map logic circuits with interconnected complexity and logic events of c-Met recognition and dimerization on cell surface for signaling regulation by MS interrogation.
Collapse
Affiliation(s)
- Xue Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| | - Yuxiang Dong
- School of Materials Science and Engineering, Suzhou University of Science and Technology, Suzhou, 215009, P. R. China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| | - Zhenzhen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| | - Xuemeng Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| | - Ye Tian
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| | - Qianhao Min
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
2
|
Sang D, Luo X, Liu J. Biological Interaction and Imaging of Ultrasmall Gold Nanoparticles. NANO-MICRO LETTERS 2023; 16:44. [PMID: 38047998 PMCID: PMC10695915 DOI: 10.1007/s40820-023-01266-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023]
Abstract
The ultrasmall gold nanoparticles (AuNPs), serving as a bridge between small molecules and traditional inorganic nanoparticles, create significant opportunities to address many challenges in the health field. This review discusses the recent advances in the biological interactions and imaging of ultrasmall AuNPs. The challenges and the future development directions of the ultrasmall AuNPs are presented.
Collapse
Affiliation(s)
- Dongmiao Sang
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, People's Republic of China
| | - Xiaoxi Luo
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, People's Republic of China
| | - Jinbin Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, People's Republic of China.
| |
Collapse
|
3
|
Jantarat T, Lauterbach JD, Doungchawee J, Agrohia DK, Vachet RW. Quantitative imaging of the sub-organ distributions of nanomaterials in biological tissues via laser ablation inductively coupled plasma mass spectrometry. Analyst 2023; 148:4479-4488. [PMID: 37575048 DOI: 10.1039/d3an00839h] [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: 08/15/2023]
Abstract
Nanomaterials have been employed in many biomedical applications, and their distributions in biological systems can provide an understanding of their behavior in vivo. Laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) can be used to determine the distributions of metal-based NMs in biological systems. However, LA-ICP-MS has not commonly been used to quantitatively measure the cell-specific or sub-organ distributions of nanomaterials in tissues. Here, we describe a new platform that uses spiked gelatin standards with control tissues on top to obtain an almost perfect tissue mimic for quantitative imaging purposes. In our approach, gelatin is spiked with both nanomaterial standards and an internal standard to improve quantitation and image quality. The value of the developed approach is illustrated by determining the sub-organ distributions of different metal-based and metal-tagged polymeric nanomaterials in mice organs. The LA-ICP-MS images reveal that the chemical and physical properties of the nanomaterials cause them to distribute in quantitatively different extents in spleens, kidneys, and tumors, providing new insight into the fate of nanomaterials in vivo. Furthermore, we demonstrate that this approach enables quantitative co-localization of nanomaterials and their cargo. We envision this method being a valuable tool in the development of nanomaterial drug delivery systems.
Collapse
Affiliation(s)
- Teerapong Jantarat
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01002, USA.
| | - Joshua D Lauterbach
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01002, USA.
| | - Jeerapat Doungchawee
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01002, USA.
| | - Dheeraj K Agrohia
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01002, USA.
| | - Richard W Vachet
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01002, USA.
| |
Collapse
|
4
|
Zhang P, Ye G, Xie G, Lv J, Zeng X, Jiang W. Research progress of nanomaterial drug delivery in tumor targeted therapy. Front Bioeng Biotechnol 2023; 11:1240529. [PMID: 37555076 PMCID: PMC10405625 DOI: 10.3389/fbioe.2023.1240529] [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: 06/15/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023] Open
Abstract
Cancer is one of the most lethal diseases in human society, and its incidence is gradually increasing. However, the current tumor treatment often meets the problem of poor efficacy and big side effects. The unique physical and chemical properties of nanomaterials can target the delivery of drugs to tumors, which can improve the therapeutic effect while reducing the damage of drugs to normal cells. This makes nanomaterials become a hot topic in the field of biomedicine. This review summarizes the recent progress of nanomaterials in tumor targeted therapy.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, China
| | - Guihua Ye
- Shanghai Ninth People’s Hospital Hainan Branch, Hainan Western Central Hospital, Danzhou, China
| | - Guofeng Xie
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Jie Lv
- School of Computer Science and Engineering, Yulin Normal University, Yulin, China
| | - Xianhai Zeng
- Department of Otorhinolaryngology, Longgang Otorhinolaryngology Hospital & Shenzhen Key Laboratory of Otorhinolaryngology, Shenzhen Institute of Otorhinolaryngology, Shenzhen, China
| | - Wei Jiang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
5
|
Prysiazhnyi V, Bednařík A, Žalud M, Hegrová V, Neuman J, Preisler J. Fate of Gold Nanoparticles in Laser Desorption/Ionization Mass Spectrometry: Toward the Imaging of Individual Nanoparticles. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:570-578. [PMID: 36917818 PMCID: PMC10080673 DOI: 10.1021/jasms.2c00300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/13/2023] [Accepted: 03/02/2023] [Indexed: 06/14/2023]
Abstract
This study focuses on mapping the spatial distribution of Au nanoparticles (NPs) by laser desorption/ionization mass spectrometry imaging (LDI MSI). Laser interaction with NPs and associated phenomena, such as change of shape, melting, migration, and release of Au ions, are explored at the single particle level. Arrays of dried droplets containing low numbers of spatially segregated NPs were reproducibly prepared by automated drop-on-demand piezo-dispensing and analyzed by LDI MSI using an ultrahigh resolution orbital trapping instrument. To enhance the signal from NPs, an in source gas-phase chemical reaction of generated Au ions with xylene was employed. The developed technique allowed the detecting, chemical characterization, and mapping of the spatial distribution of Au NPs; the ion signals were detected from as low as ten 50 nm Au NPs on a pixel. Furthermore, the Au NP melting dynamics under laser irradiation was monitored by correlative atomic force microscopy (AFM) and scanning electron microscopy (SEM). AFM measurements of Au NPs before and after LDI MSI analysis revealed changes in NP shape from a sphere to a half-ellipsoid and total volume reduction of NPs down to 45% of their initial volume.
Collapse
Affiliation(s)
- Vadym Prysiazhnyi
- Department
of Chemistry, Faculty of Science, Masaryk
University, 625 00, Brno, Czech
Republic
| | - Antonín Bednařík
- Department
of Chemistry, Faculty of Science, Masaryk
University, 625 00, Brno, Czech
Republic
| | - Michal Žalud
- Department
of Chemistry, Faculty of Science, Masaryk
University, 625 00, Brno, Czech
Republic
| | | | - Jan Neuman
- Nenovision
s. r. o., 612 00, Brno, Czech Republic
| | - Jan Preisler
- Department
of Chemistry, Faculty of Science, Masaryk
University, 625 00, Brno, Czech
Republic
| |
Collapse
|
6
|
Dominique NL, Jensen IM, Kaur G, Kotseos CQ, Boggess WC, Jenkins DM, Camden JP. Giving Gold Wings: Ultrabright and Fragmentation Free Mass Spectrometry Reporters for Barcoding, Bioconjugation Monitoring, and Data Storage. Angew Chem Int Ed Engl 2023; 62:e202219182. [PMID: 36853583 DOI: 10.1002/anie.202219182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/01/2023]
Abstract
The widespread application of laser desorption/ionization mass spectrometry (LDI-MS) highlights the need for a bright and multiplexable labeling platform. While ligand-capped Au nanoparticles (AuNPs) have emerged as a promising LDI-MS contrast agent, the predominant thiol ligands suffer from low ion yields and extensive fragmentation. In this work, we develop a N-heterocyclic carbene (NHC) ligand platform that enhances AuNP LDI-MS performance. NHC scaffolds are tuned to generate barcoded AuNPs which, when benchmarked against thiol-AuNPs, are bright mass tags and form unfragmented ions in high yield. To illustrate the transformative potential of NHC ligands, the mass tags were employed in three orthogonal applications: monitoring a bioconjugation reaction, performing multiplexed imaging, and storing and reading encoded information. These results demonstrate that NHC-nanoparticle systems are an ideal platform for LDI-MS and greatly broaden the scope of nanoparticle contrast agents.
Collapse
Affiliation(s)
- Nathaniel L Dominique
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - Isabel M Jensen
- Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, 37996, USA
| | - Gurkiran Kaur
- Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, 37996, USA
| | - Chandler Q Kotseos
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - William C Boggess
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - David M Jenkins
- Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, 37996, USA
| | - Jon P Camden
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, 46556, USA
| |
Collapse
|
7
|
Yin H, Chu Y, Wang W, Zhang Z, Meng Z, Min Q. Mass tag-encoded nanointerfaces for multiplexed mass spectrometric analysis and imaging of biomolecules. NANOSCALE 2023; 15:2529-2540. [PMID: 36688447 DOI: 10.1039/d2nr06020e] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Revealing multiple biomolecules in the physiopathological environment simultaneously is crucial in biological and biomedical research. Mass spectrometry (MS) features unique technical advantages in multiplexed and label-free analyses. However, owing to comparably low abundance and poor ionization efficiency of target biomolecules, direct MS profiling of these biological species in vitro or in situ remains a challenge. An emerging route to solve this issue is to devise mass tag (MT)-encoded nanointerfaces which specifically convert the abundance or activity of biomolecules into amplified ion signals of mass tags, offering an ideal strategy for synchronous MS assaying and mapping of multiple targets in biofluids, cells and tissues. This review provides a thorough and organized overview of recent advances in MT-encoded nanointerfaces elaborately tailored for several practical applications in multiplexed MS bioanalysis and biomedical research. First, we start with elucidation of the structural characteristics and working principle of MT-encoded nanointerfaces in specific labeling and sensing of multiple biological targets. In addition, we further discuss the application scenarios of MT-encoded nanointerfaces particularly in multiplexed biomarker assays, cell analysis, and tissue imaging. Finally, the current challenges are pointed out and future prospects of these nanointerfaces in MS analysis are forecast.
Collapse
Affiliation(s)
- Hao Yin
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Yanxin Chu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Wei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Zhenzhen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Zhen Meng
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Qianhao Min
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| |
Collapse
|
8
|
Su Y, Lai X, Guo K, Wang X, Chen S, Liang K, Pu K, Wang Y, Hu J, Wei X, Chen Y, Wang H, Lin W, Ni W, Lin Y, Zhu J, Ng KM. Covalent Bonding and Coulomb Repulsion-Guided AuNP Array: A Tunable and Reusable Substrate for Metabolomic Characterization of Lung Cancer Patient Sera. Anal Chem 2022; 94:16910-16918. [PMID: 36417775 DOI: 10.1021/acs.analchem.2c04319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Surface-assisted laser desorption/ionization mass spectrometry (SALDI-MS) has gained increased attention in the metabolic characterization of human biofluids. However, the stability and reproducibility of nanoparticle-based substrates remain two of the biggest challenges in high-salt environments. Here, by controlling the extent of Coulomb repulsion of 26 nm positively charged AuNPs, a homogeneous layer of covalently bonded AuNPs on a coverslip with tunable interparticle distances down to 16 nm has been successfully fabricated to analyze small biomolecules in human serum. Compared with the self-assembled AuNP array, the covalently bonded AuNP array showed superior performances on stability, reproducibility, and sensitivity in high-salt environments. The stable attachment of AuNPs maintained a detection reproducibility with a RSD less than 12% and enabled the reusability of the array for 10 experiments without significant signal deterioration (<15%) and carryover effects. Moreover, the closely positioned AuNPs allowed the coupling of photoinduced plasmons to generate an enhanced electric field, which promotes the generation of excited electrons to facilitate the desorption/ionization processes instead of the heat dissipation, thus enhancing the detection sensitivity with detection limits down to the femtomole level. Combined with machine learning methods, the AuNP array has been successfully applied to discover seven biomarkers for differentiating early-stage lung cancer patients from healthy controls. It is anticipated that this simple approach of developing robust AuNP arrays can also be extended to other types of NP arrays for wider applications of SALDI-MS technology.
Collapse
Affiliation(s)
- Yang Su
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, Shantou, Guangdong 515063, P. R. China
| | - Xiaopin Lai
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, Shantou, Guangdong 515063, P. R. China
| | - Kunbin Guo
- The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Xin Wang
- The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Siyu Chen
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, Shantou, Guangdong 515063, P. R. China
| | - Kaiqing Liang
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, Shantou, Guangdong 515063, P. R. China
| | - Keyuan Pu
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, Shantou, Guangdong 515063, P. R. China
| | - Yue Wang
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, Shantou, Guangdong 515063, P. R. China
| | - Jun Hu
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, Shantou, Guangdong 515063, P. R. China
| | - Xiaolong Wei
- The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Yuping Chen
- The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Hongbiao Wang
- The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Wen Lin
- The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Wenxiu Ni
- Department of Medicinal Chemistry, Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Yan Lin
- The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Janshon Zhu
- Guangdong RangerBio Technologies Company Limited, Dongguan 523000, P. R. China
| | - Kwan-Ming Ng
- Department of Chemistry and Key Laboratory for Preparation and Application of Ordered Structural Materials of Guangdong Province, Shantou University, Shantou, Guangdong 515063, P. R. China
| |
Collapse
|
9
|
Nanomaterial characterization: Understanding nano-bio interactions. Biochem Biophys Res Commun 2022; 633:45-51. [DOI: 10.1016/j.bbrc.2022.08.095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 08/31/2022] [Indexed: 11/06/2022]
|
10
|
Agrohia DK, Wu P, Huynh U, Thayumanavan S, Vachet RW. Multiplexed Analysis of the Cellular Uptake of Polymeric Nanocarriers. Anal Chem 2022; 94:7901-7908. [PMID: 35612963 DOI: 10.1021/acs.analchem.2c00648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Polymeric nanocarriers (PNCs) are versatile drug delivery vehicles capable of delivering a variety of therapeutics. Quantitatively monitoring their uptake in biological systems is essential for realizing their potential as next-generation delivery systems; however, existing quantification strategies are limited due to the challenges of detecting polymeric materials in complex biological samples. Here, we describe a metal-coded mass tagging approach that enables the multiplexed quantification of the PNC uptake in cells using mass spectrometry (MS). In this approach, PNCs are conjugated with ligands that bind strongly to lanthanide ions, allowing the PNCs to be sensitively quantitated by inductively coupled plasma-MS. The metal-coded tags have little effect on the properties or toxicity of the PNCs, making them biocompatible. We demonstrate that the conjugation of different metals to the PNCs enables the multiplexed analysis of cellular uptake of multiple distinct PNCs at the same time. This multiplexing capability should improve the design and optimization of PNCs by minimizing biological variability and reducing analysis time, effort, and cost.
Collapse
Affiliation(s)
- Dheeraj K Agrohia
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Peidong Wu
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Uyen Huynh
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - S Thayumanavan
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Center for Bioactive Delivery─Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| | - Richard W Vachet
- Department of Chemistry, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Molecular and Cellular Biology Program, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States.,Center for Bioactive Delivery─Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts 01003, United States
| |
Collapse
|
11
|
Loutfy SA, Abdel-Salam AI, Moatasim Y, Gomaa MR, Abdel Fattah NF, Emam MH, Ali F, ElShehaby HA, Ragab EA, Alam El-Din HM, Mostafa A, Ali MA, Kasry A. Antiviral activity of chitosan nanoparticles encapsulating silymarin (Sil-CNPs) against SARS-CoV-2 ( in silico and in vitro study). RSC Adv 2022; 12:15775-15786. [PMID: 35685696 PMCID: PMC9132606 DOI: 10.1039/d2ra00905f] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/19/2022] [Indexed: 02/06/2023] Open
Abstract
To develop a specific treatment against COVID-19, we investigated silymarin-chitosan nanoparticles (Sil-CNPs) as an antiviral agent against SARS-CoV-2 using in silico and in vitro approaches. Docking of Sil and CNPs was carried out against SARS-CoV-2 spike protein using AutoDock Vina. CNPs and Sil-CNPs were prepared by the ionic gelation method and characterized by TEM, FT-IR, zeta analysis, and the membrane diffusion method to determine the drug release profile. Cytotoxicity was tested on both Vero and Vero E6 cell lines using the MTT assay. Minimum binding energies with spike protein and ACE2 were -6.6, and -8.0 kcal mol-1 for CNPs, and -8.9, and -9.7 kcal mol-1 for Sil, respectively, compared to -6.6 and -8.4 kcal mol-1 respectively for remdesivir (RMV). CNPs and Sil-CNPs were prepared at sizes of 29 nm and 82 nm. The CC50 was 135, 35, and 110 μg mL-1 for CNPs, Sil, and Sil-CNPs, respectively, on Vero E6. The IC50 was determined at concentrations of 0.9, 12 and 0.8 μg mL-1 in virucidal/replication assays for CNPs, Sil, and Sil-CNPs respectively using crystal violet. These results indicate antiviral activity of Sil-CNPs against SARS-CoV-2.
Collapse
Affiliation(s)
- Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute (NCI), Cairo University Fom El-Khalig 11796 Cairo Egypt
- Nanotechnology Research Center (NTRC), The British University in Egypt El-Shorouk City, Suez Desert Road P. O. Box 43 Cairo 11837 Egypt
| | - Ahmed I Abdel-Salam
- Nanotechnology Research Center (NTRC), The British University in Egypt El-Shorouk City, Suez Desert Road P. O. Box 43 Cairo 11837 Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre (NRC) Giza 12622 Egypt
| | - Mokhtar R Gomaa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre (NRC) Giza 12622 Egypt
| | - Nasra F Abdel Fattah
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute (NCI), Cairo University Fom El-Khalig 11796 Cairo Egypt
| | - Merna H Emam
- Nanotechnology Research Center (NTRC), The British University in Egypt El-Shorouk City, Suez Desert Road P. O. Box 43 Cairo 11837 Egypt
| | - Fedaa Ali
- Nanotechnology Research Center (NTRC), The British University in Egypt El-Shorouk City, Suez Desert Road P. O. Box 43 Cairo 11837 Egypt
| | | | - Eman A Ragab
- Biochemistry Dept, Faculty of Science, Cairo University Egypt
| | - Hanaa M Alam El-Din
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute (NCI), Cairo University Fom El-Khalig 11796 Cairo Egypt
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre (NRC) Giza 12622 Egypt
| | - Mohamed A Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre (NRC) Giza 12622 Egypt
| | - Amal Kasry
- Nanotechnology Research Center (NTRC), The British University in Egypt El-Shorouk City, Suez Desert Road P. O. Box 43 Cairo 11837 Egypt
| |
Collapse
|
12
|
Jakubczak W, Haczyk-Więcek M, Pawlak K. Attomole-per Cell Atomic Mass Spectrometry Measurement of Platinum and Gold Drugs in Cultured Lung Cancer Cells. Molecules 2021; 26:7627. [PMID: 34946708 PMCID: PMC8703441 DOI: 10.3390/molecules26247627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/06/2021] [Accepted: 12/13/2021] [Indexed: 12/22/2022] Open
Abstract
In this study, we developed a strategy to determine atto- and femtomolar amounts of metal ions in lysates and mineralizates of cells (human non-small-cell lung carcinoma (NSCLC, A549) and normal lung (MRC-5)) exposed to cytotoxic metallo-drugs: cisplatin and auranofin at concentrations close to the half-maximal inhibitory drug concentrations (IC50). The developed strategy combines data obtained using biological and chemical approaches. Cell density was determined using two independent cell staining assays using trypan blue, calcein AM/propidium iodide. Metal concentrations in lysed and mineralized cells were established employing a mass spectrometer with inductively coupled plasma (ICP-MS) and equipped with a cross-flow nebulizer working in aspiration mode. It allowed for detecting of less than 1 fg of metal per cell. To decrease the required amount of sample material (from 1.5 mL to ~100 µL) without loss of sensitivity, the sample was introduced as a narrow band into a constant stream of liquid (flow-injection analysis). It was noticed that the selectivity of cisplatin accumulation by cells depends on the incubation time. This complex is accumulated by cells at a lower efficiency than auranofin and is found primarily in the lysate representing the cytosol. In contrast, auranofin interacts with water-insoluble compounds. Despite their different mechanism of action, both metallo-drugs increased the accumulation of transition metal ions responsible for oxidative stress.
Collapse
Affiliation(s)
| | | | - Katarzyna Pawlak
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland; (W.J.); (M.H.-W.)
| |
Collapse
|
13
|
Xu H, Zhang Z, Wang Y, Lu W, Min Q. Engineering of nanomaterials for mass spectrometry analysis of biomolecules. Analyst 2021; 146:5779-5799. [PMID: 34397044 DOI: 10.1039/d1an00860a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mass spectrometry (MS) based analysis has received intense attention in diverse biological fields. However, direct MS interrogation of target biomolecules in complex biological samples is still challenging, due to the extremely low abundance and poor ionization potency of target biological species. Innovations in nanomaterials create new auxiliary tools for deep and comprehensive MS characterization of biomolecules. More recently, growing research interest has been directed to the compositional and structural engineering of nanomaterials for enriching target biomolecules prior to MS analysis, enhancing the ionization efficiency in MS detection and designing biosensing nanoprobes in sensitive MS readout. In this review, we mainly focus on the recent advances in the engineering of nanomaterials towards their applications in sample pre-treatment, desorption/ionization matrices and ion signal amplification for MS profiling of biomolecules. This review will provide a toolbox of nanomaterials for researchers devoted to developing analytical methods and practical applications in the biological MS field.
Collapse
Affiliation(s)
- Hongmei Xu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China. and Institute of Environmental Science, Shanxi University, Taiyuan 030006, P. R. China
| | - Zhenzhen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Yihan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Weifeng Lu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| | - Qianhao Min
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China.
| |
Collapse
|
14
|
Lima AC, Reis RL, Ferreira H, Neves NM. Cellular Uptake of Three Different Nanoparticles in an Inflammatory Arthritis Scenario versus Normal Conditions. Mol Pharm 2021; 18:3235-3246. [PMID: 34387081 DOI: 10.1021/acs.molpharmaceut.1c00066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nanoparticles (NPs) have wide potential applications in the biomedical field. To promote targeted and controlled delivery of encapsulated drugs, it is fundamentally important to understand the factors regulating NP uptake by different cells. Thus, the goal of the present study is to assess the internalization rates of different NPs under normal and proinflammatory states in primary human articular chondrocytes (hACs), human umbilical vein endothelial cells (EA), and human monocytes (THP-1). Here, we compared chitosan-hyaluronic acid (Ch-HA) polymeric NPs, methoxypolyethylene glycol amine-glutathione-palmitic acid (mPEG-GSHn-PA) micelles, and cholesterol/l-α-phosphatidylcholine/DSPE-PEG-Mal (Chol/EPC/DSPE-PEG-Mal) unilamellar liposomes (LUVs). Our results reveal the importance of surface charge and chemistry in determining the levels of NP internalization. Under normal conditions, the cellular uptake was ≈30% for Ch-HA NPs and ≈100% for mPEG-GSHn-PA micelles and Chol/EPC/DSPE-PEG-Mal LUVs. A proinflammatory cell state promoted a higher uptake of the Ch-HA NPs by EA cells (93% after 24 h). Since the therapeutic efficacy of the NP-loaded cargo is dependent on trafficking routes after cellular internalization, we tested their internalization pathways. Accordingly, caveolae-mediated endocytosis or energy-independent non-endocytic pathways, which circumvent lysosomal degradation, were accomplished in hACs and EA by LUVs and in M1 polarized macrophages by micelles. The present outcomes highlight the importance of considering cellular uptake and internalization pathways by the target cell when designing functional NPs for therapeutic applications.
Collapse
Affiliation(s)
- Ana Cláudia Lima
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Helena Ferreira
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Nuno M Neves
- 3B's Research Group-Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4805-017 Barco, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
15
|
Huang R, Luther DC, Zhang X, Gupta A, Tufts SA, Rotello VM. Engineering the Interface between Inorganic Nanoparticles and Biological Systems through Ligand Design. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1001. [PMID: 33924735 PMCID: PMC8069843 DOI: 10.3390/nano11041001] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/15/2022]
Abstract
Nanoparticles (NPs) provide multipurpose platforms for a wide range of biological applications. These applications are enabled through molecular design of surface coverages, modulating NP interactions with biosystems. In this review, we highlight approaches to functionalize nanoparticles with "small" organic ligands (Mw < 1000), providing insight into how organic synthesis can be used to engineer NPs for nanobiology and nanomedicine.
Collapse
Affiliation(s)
| | | | | | | | | | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 N. Pleasant St., Amherst, MA 01003, USA; (R.H.); (D.C.L.); (X.Z.); (A.G.); (S.A.T.)
| |
Collapse
|
16
|
Geng Y, Amante JJ, Goel HL, Zhang X, Walker MR, Luther DC, Mercurio AM, Rotello VM. Differentiation of Cancer Stem Cells through Nanoparticle Surface Engineering. ACS NANO 2020; 14:15276-15285. [PMID: 33164505 PMCID: PMC10566532 DOI: 10.1021/acsnano.0c05589] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cancer stem cells (CSCs) are a crucial therapeutic target because of their role in resistance to chemo- and radiation therapy, metastasis, and tumor recurrence. Differentiation therapy presents a potential strategy for "defanging" CSCs. To date, only a limited number of small-molecule and nanomaterial-based differentiating agents have been identified. We report here the integrated use of nanoparticle engineering and hypothesis-free sensing to identify nanoparticles capable of efficient differentiation of CSCs into non-CSC phenotypes. Using this strategy, we identified a nanoparticle that induces CSC differentiation by increasing intracellular reactive oxygen species levels. Importantly, this unreported phenotype is more susceptible to drug treatment than either CSCs or non-CSCs, demonstrating a potentially powerful strategy for anticancer therapeutics.
Collapse
Affiliation(s)
- Yingying Geng
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, United States
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, United States
| | - John J. Amante
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Hira L. Goel
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Xianzhi Zhang
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, United States
| | - Melanie R. Walker
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - David C. Luther
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, United States
| | - Arthur M. Mercurio
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, United States
| | - Vincent M. Rotello
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01003, United States
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, United States
| |
Collapse
|
17
|
Xu H, Huang X, Zhang Z, Zhang X, Min Q, Zhu JJ. Protease-responsive mass barcoded nanotranslators for simultaneously quantifying the intracellular activity of cascaded caspases in apoptosis pathways. Chem Sci 2020; 11:5280-5288. [PMID: 34122985 PMCID: PMC8159337 DOI: 10.1039/d0sc01534b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Quantitatively delineating the activation network of multiple proteases that participate in cellular processes is highly essential for understanding the physiological and pathological states of cells. In this study, protease-responsive mass barcoded nanotranslators (PRMNTs) were engineered for revealing the activity of cascaded caspases in apoptosis in a multiplex and quantitative manner. In the PRMNTs, a series of mass tag-decorated gold nanoparticles (AuNPs) were tethered onto magnetic Fe3O4 nanospheres via a linker containing the substrate peptide of the target protease to form a "one-to-many" core-satellite structure. This nanostructure was internalized into the cells, underwent an enzymatic reaction within the cells, and allowed post-reaction mass spectrometry (MS) interrogation after magnetic separation from the cells. In the presence of intracellular caspases, enzymatic cleavage of the linker could be translated to the decreased ion signals of the mass tags on the remaining AuNPs in the PRMNTs by MS decoding. Benefiting from the multiplexing capability of MS, the intracellular activity of caspase-3, -8 and -9 that orchestrate the apoptotic process was simultaneously quantified at any given time. Kinetic analysis of caspase activity under stimulation of diverse anticancer drugs revealed that programmed cell death followed individual apoptosis pathways, differing in the activation degree and sequence of the caspase cascade. This work represents a modality that interfaces nanotechnology with MS for quantitatively probing the intracellular activity of multiple proteases, which opens up new avenues for revealing the apoptosis mechanism and developing innovative drugs.
Collapse
Affiliation(s)
- Hongmei Xu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
| | - Xiaodan Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
| | - Zhenzhen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
| | - Xuemeng Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
| | - Qianhao Min
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science, Chemistry and Biomedicine Innovation Center, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 P. R. China
| |
Collapse
|
18
|
Loutfy SA, Elberry MH, Farroh KY, Mohamed HT, Mohamed AA, Mohamed EB, Faraag AHI, Mousa SA. Antiviral Activity of Chitosan Nanoparticles Encapsulating Curcumin Against Hepatitis C Virus Genotype 4a in Human Hepatoma Cell Lines. Int J Nanomedicine 2020; 15:2699-2715. [PMID: 32368050 PMCID: PMC7184126 DOI: 10.2147/ijn.s241702] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/23/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose Current direct-acting antiviral agents for treatment of hepatitis C virus genotype 4a (HCV-4a) have been reported to cause adverse effects, and therefore less toxic antivirals are needed. This study investigated the role of curcumin chitosan (CuCs) nanocomposite as a potential anti-HCV-4a agent in human hepatoma cells Huh7. Methods Docking of curcumin and CuCs nanocomposite and binding energy calculations were carried out. Chitosan nanoparticles (CsNPs) and CuCs nanocomposite were prepared with an ionic gelation method and characterized with TEM, zeta size and potential, and HPLC to calculate encapsulation efficiency. Cytotoxicity studies were performed on Huh7 cells using MTT assay and confirmed with cellular and molecular assays. Anti-HCV-4a activity was determined using real-time PCR and Western blot. Results The strength of binding interactions between protein ligand complexes gave scores with NS3 protease, NS5A polymerase, and NS5B polymerase of -124.91, -159.02, and -129.16, for curcumin respectively, and -68.51, -54.52, and -157.63 for CuCs nanocomposite, respectively. CuCs nanocomposite was prepared at sizes 29-39.5 nm and charges of 33 mV. HPLC detected 4% of curcumin encapsulated into CsNPs. IC50 was 8 µg/mL for curcumin and 25 µg/mL for the nanocomposite on Huh7 but was 25.8 µg/mL and 34 µg/mL on WISH cells. CsNPs had no cytotoxic effect on tested cell lines. Apoptotic genes' expression revealed the caspase-dependent pathway mechanism. CsNPs and CuCs nanocomposite demonstrated 100% inhibition of viral entry and replication, which was confirmed with HCV core protein expression. Conclusion CuCs nanocomposite inhibited HCV-4a entry and replication compared to curcumin alone, suggesting its potential role as an effective therapeutic agent.
Collapse
Affiliation(s)
- Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.,Nanotechnology Research Center, British University, Cairo, Egypt
| | - Mostafa H Elberry
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Khaled Yehia Farroh
- Nanotechnology and Advanced Materials Central Lab, Agricultural Research Center, Giza, Egypt
| | - Hossam Taha Mohamed
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th October, Giza, Egypt.,Department of Zoology, Faculty of Science,Cairo University, Giza, Egypt
| | - Aya A Mohamed
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th October, Giza, Egypt
| | - ElChaimaa B Mohamed
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Ahmed Hassan Ibrahim Faraag
- Botany and Microbiology Department, Bioinformatics Center, Faculty of Science, Helwan University, Cairo, Egypt
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| |
Collapse
|
19
|
Le Goas M, Roussel T, Kalbazova M, Carrière D, Barruet E, Geertsen V, Fadda GC, Testard F, Carrot G, Renault JP. Combining surface chemistry modification and in situ small-angle scattering characterization to understand and optimize the biological behavior of nanomedicines. J Mater Chem B 2020; 8:6438-6450. [DOI: 10.1039/d0tb01167c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Monitoring structural transformation and quantity of NPs in biologically relevant environments with small-angle scattering techniques.
Collapse
Affiliation(s)
| | - Tom Roussel
- Université Paris-Saclay
- CEA
- CNRS
- NIMBE
- Gif-sur-Yvette
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Han J, Huang X, Liu H, Wang J, Xiong C, Nie Z. Laser cleavable probes for in situ multiplexed glycan detection by single cell mass spectrometry. Chem Sci 2019; 10:10958-10962. [PMID: 32190253 PMCID: PMC7066667 DOI: 10.1039/c9sc03912k] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
A single-cell MS approach for multiplexed glycan detection to investigate the relationship between drug resistance and glycans at a single-cell level and quantify multiple glycans, overcoming the limit of low ionization efficiency of glycans.
Glycans binding on the cell surface through glycosylation play a key role in controlling various cellular processes, and glycan analysis at a single-cell level is necessary to study cellular heterogeneity and diagnose diseases in the early stage. Herein, we synthesized a series of laser cleavable probes, which could sensitively detect glycans on single cells and tissues by laser desorption ionization mass spectrometry (LDI-MS). This multiplexed and quantitative glycan detection was applied to evaluate the alterations of four types of glycans on breast cancer cells and drug-resistant cancer cells at a single-cell level, indicating that drug resistance may be related to the upregulation of glycan with a β-d-galactoside (Galβ) group and Neu5Aca2-6Gal(NAc)-R. Moreover, the glycan spatial distribution in cancerous and paracancerous human tissues was also demonstrated by MS imaging, showing that glycans are overexpressed in cancerous tissues. Therefore, this single-cell MS approach exhibits promise for application in studying glycan functions which are essential for clinical biomarker discovery and diagnosis of related diseases.
Collapse
Affiliation(s)
- Jing Han
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China . ; .,University of the Chinese Academy of Sciences , Beijing 100049 , China
| | - Xi Huang
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China . ;
| | - Huihui Liu
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China . ;
| | - Jiyun Wang
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China . ;
| | - Caiqiao Xiong
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China . ;
| | - Zongxiu Nie
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , China . ; .,University of the Chinese Academy of Sciences , Beijing 100049 , China.,National Center for Mass Spectrometry in Beijing , Beijing 100049 , China
| |
Collapse
|
21
|
Malile B, Brkic J, Bouzekri A, Wilson DJ, Ornatsky O, Peng C, Chen JIL. DNA-Conjugated Gold Nanoparticles as High-Mass Probes in Imaging Mass Cytometry. ACS APPLIED BIO MATERIALS 2019; 2:4316-4323. [DOI: 10.1021/acsabm.9b00574] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Brian Malile
- Department of Chemistry, York University, 4700 Keele Street, Toronto, Ontario, Canada M3J 1P3
| | - Jelena Brkic
- Department of Biology, York University, 4700 Keele Street, Toronto, Ontario, Canada M3J 1P3
| | - Alexandre Bouzekri
- Fluidigm Canada Inc., 1380 Rodick Road, Markham, Ontario, Canada L3R 4G5
| | - Derek J. Wilson
- Department of Chemistry, York University, 4700 Keele Street, Toronto, Ontario, Canada M3J 1P3
- Centre for Research in Mass Spectrometry, York University, 4700 Keele Street, Toronto, Ontario, Canada M3J 1P3
- Centre for Research on Biomolecular Interactions, York University, 4700 Keele Street, Toronto, Ontario, Canada M3J 1P3
| | - Olga Ornatsky
- Fluidigm Canada Inc., 1380 Rodick Road, Markham, Ontario, Canada L3R 4G5
| | - Chun Peng
- Department of Biology, York University, 4700 Keele Street, Toronto, Ontario, Canada M3J 1P3
- Centre for Research on Biomolecular Interactions, York University, 4700 Keele Street, Toronto, Ontario, Canada M3J 1P3
| | - Jennifer I. L. Chen
- Department of Chemistry, York University, 4700 Keele Street, Toronto, Ontario, Canada M3J 1P3
| |
Collapse
|
22
|
Aghamiri S, Mehrjardi KF, Shabani S, Keshavarz-Fathi M, Kargar S, Rezaei N. Nanoparticle-siRNA: a potential strategy for ovarian cancer therapy? Nanomedicine (Lond) 2019; 14:2083-2100. [PMID: 31368405 DOI: 10.2217/nnm-2018-0379] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer is one of the most common causes of mortality throughout the world. Unfortunately, chemotherapy has failed to cure advanced cancers developing multidrug resistance (MDR). Moreover, it has critical side effects because of nonspecific toxicity. Thanks to specific silencing of oncogenes and MDR-associated genes, nano-siRNA drugs can be a great help address the limitations of chemotherapy. Here, we review the current advances in nanoparticle-mediated siRNA delivery strategies such as polymeric- and lipid-based systems, rigid nanoparticles and nanoparticles coupled to specific ligand systems. Nanoparticle-based codelivery of anticancer drugs and siRNA targeting various mechanisms of MDR is a cutting-edge strategy for ovarian cancer therapy, which is completely discussed in this review.
Collapse
Affiliation(s)
- Shahin Aghamiri
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technology in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 19839-63113, Iran
| | - Keyvan Fallah Mehrjardi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran
| | - Sasan Shabani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education & Research Network (USERN), Tehran, 1419733151, Iran.,Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Saeed Kargar
- Department of Life Science Engineering, Faculty of New Sciences & Technologies, University of Tehran, Tehran, 1417466191, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, 1419733151, Iran
| |
Collapse
|
23
|
Salaheldin TA, Loutfy SA, Ramadan MA, Youssef T, Mousa SA. IR-enhanced photothermal therapeutic effect of graphene magnetite nanocomposite on human liver cancer HepG2 cell model. Int J Nanomedicine 2019; 14:4397-4412. [PMID: 31417251 PMCID: PMC6592061 DOI: 10.2147/ijn.s196256] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 04/16/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Graphene magnetite nanocomposites (G/Fe3O4) exhibit light photothermal conversion upon enhancement by 808 nm IR laser excitation. We evaluated the cytotoxic and photothermal effects of G/Fe3O4 on a HepG2 human liver cancer cell model. Methods: Graphene nanosheets (rGO), magnetite nanoparticles (Fe3O4), and G/Fe3O4 were prepared by chemical methods and characterized using transmission electron microscopy, Raman spectroscopy, zeta analysis, and vibrating sample magnemeter. Dark and light cytotoxicity were screened with colorimetric Sulforhodamine B cell viability assay after 24 and 48 hours. DNA fragmentation and some apoptotic genes on a transcriptional RNA level expression were performed. All prepared nanomaterials were evaluated for their photothermal effect at concentrations of 10 and 50 µg/mL. The power density incident on the cells by 300 mW 808 IR diode laser was 0.597 W/cm2. Results: Treatment of HepG2 with 400 µg/mL of rGO, Fe3O4, and G/Fe3O4 showed alteration in cell morphology after 24 hours of cell treatment and revealed toxic effects on cellular DNA. Evaluation of the cytotoxic effects showed messenger RNA (mRNA) in β-actin and Bax apoptotic genes, but no expression of mRNA of caspase-3 after 24 hours of cell exposure, suggesting the involvement of an intrinsic apoptotic caspase-independent pathway. A photothermal effect was observed for G/Fe3O4 after irradiation of the HepG2 cells. A marked decrease was found in cell viability when treated with 10 and 50 µg/mL G/Fe3O4 from 40% to 5% after 48 hours of cell treatment. Conclusion: Results indicate that G/Fe3O4 nanocomposite was effective at transformation of light into heat and is a promising candidate for cancer therapy.
Collapse
Affiliation(s)
- Taher A Salaheldin
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
- Nanotechnology and Advanced Materials Central Lab, Agricultural Research Center, Giza, Egypt
| | - Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Marwa A Ramadan
- Department of Photochemistry Photobiology, National Institute for Laser Enhanced Science (NILES) Cairo University, Cairo, Egypt
| | - Tareq Youssef
- Department of Photochemistry Photobiology, National Institute for Laser Enhanced Science (NILES) Cairo University, Cairo, Egypt
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| |
Collapse
|
24
|
Ma W, Xu S, Nie H, Hu B, Bai Y, Liu H. Bifunctional cleavable probes for in situ multiplexed glycan detection and imaging using mass spectrometry. Chem Sci 2019; 10:2320-2325. [PMID: 30881658 PMCID: PMC6385553 DOI: 10.1039/c8sc04642e] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 12/21/2018] [Indexed: 11/21/2022] Open
Abstract
In situ analysis of glycans is of great significance since they mediate a range of biological activities. Aberrant changes of glycosylation are closely related to cancer onset and progression. In this work, bifunctional laser cleavable mass probes (LCMPs) were developed for in situ glycan detection from both cells and tissues using laser desorption ionization mass spectrometry (LDI-MS). Specific recognition of glycans was achieved by lectins, and inherent signal amplification was achieved by the conversion of the detection of glycans to that of mass tags which overcame the low ionization efficiency and complicated mass spectra of glycans. Multiplexed glycan profiling was easy to implement due to the simple and generic synthetic route to LCMPs and serial alternative mass tags, which offers high sensitivity, low interference and in situ detection of glycans. Moreover, as an excellent inherent matrix, LCMPs facilitated direct glycan detection from the cell surface and tissue imaging using LDI-MS. Intrinsic and fine glycan distribution in human cancer and paracancerous tissues was strictly demonstrated by MS imaging to explore the correlation between glycosylation and various cancers. This approach presented a versatile LDI-MS based platform for fast and in situ multiplexed glycan engineering, thus providing a new perspective in glycobiology and clinical diagnosis.
Collapse
Affiliation(s)
- Wen Ma
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education , College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , P. R. China . ; Tel: +86 10 6275 8198
| | - Shuting Xu
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education , College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , P. R. China . ; Tel: +86 10 6275 8198
| | - Honggang Nie
- Analytical Instrumentation Center , Peking University , Beijing , 100871 , P. R. China
| | - Bingyang Hu
- Institute of Hepatobiliary Surgery , Hospital of Hepatobiliary Surgery , Chinese People's Liberation Army General Hospital , Beijing 100853 , P. R. China
| | - Yu Bai
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education , College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , P. R. China . ; Tel: +86 10 6275 8198
| | - Huwei Liu
- Beijing National Laboratory for Molecular Sciences , Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education , College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , P. R. China . ; Tel: +86 10 6275 8198
| |
Collapse
|
25
|
Lee D, Heo DN, Nah HR, Lee SJ, Ko WK, Lee JS, Moon HJ, Bang JB, Hwang YS, Reis RL, Kwon IK. Injectable hydrogel composite containing modified gold nanoparticles: implication in bone tissue regeneration. Int J Nanomedicine 2018; 13:7019-7031. [PMID: 30464456 PMCID: PMC6219106 DOI: 10.2147/ijn.s185715] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND For effective bone regeneration, it is necessary to implant a biocompatible scaffold that is capable of inducing cell growth and continuous osteogenic stimulation at the defected site. Here, we suggest an injectable hydrogel system using enzymatic cross-linkable gelatin (Gel) and functionalized gold nanoparticles (GNPs). METHODS In this work, tyramine (Ty) was synthesized on the gelatin backbone (Gel-Ty) to enable a phenol crosslinking reaction with horseradish peroxidase (HRP). N-acetyl cysteine (NAC) was attached to the GNPs surface (G-NAC) for promoting osteodifferentiation. RESULTS The Gel-Ty hydrogels containing G-NAC (Gel-Ty/G-NAC) had suitable mechanical strength and biocompatibility to embed and support the growth of human adipose derived stem cells (hASCs) during a proliferation test for three days. In addition, G-NAC promoted osteodifferentiation both when it was included in Gel-Ty and when it was used directly in hASCs. The osteogenic effects were demonstrated by the alkaline phosphatase (ALP) activity test. CONCLUSION These findings indicate that the phenol crosslinking reaction is suitable for injectable hydrogels for tissue regeneration and G-NAC stimulate bone regeneration. Based on our results, we suggest that Gel-Ty/G-NAC hydrogels can serve both as a biodegradable graft material for bone defect treatment and as a good template for tissue engineering applications such as drug delivery, cell delivery, and various tissue regeneration uses.
Collapse
Affiliation(s)
- Donghyun Lee
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea,
| | - Dong Nyoung Heo
- Department of Engineering Science and Mechanics, Pennsylvania State University, Pennsylvania 16802, USA
| | - Ha Ram Nah
- Department of Detistry, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sang Jin Lee
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea,
| | - Wan-Kyu Ko
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si, Gyeonggi-do 13496, Republic of Korea
| | - Jae Seo Lee
- Department of Detistry, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ho-Jin Moon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea,
| | - Jae Beum Bang
- Department of Dental Education, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yu-Shik Hwang
- Department of Maxillofacial Biomedical Engineering, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Rui L Reis
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea,
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Barco, Guimarães, Portugal
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea,
| |
Collapse
|
26
|
Palermo A, Forsberg EM, Warth B, Aisporna AE, Billings E, Kuang E, Benton HP, Berry D, Siuzdak G. Fluorinated Gold Nanoparticles for Nanostructure Imaging Mass Spectrometry. ACS NANO 2018; 12:6938-6948. [PMID: 29966083 DOI: 10.1021/acsnano.8b02376] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Nanostructure imaging mass spectrometry (NIMS) with fluorinated gold nanoparticles (f-AuNPs) is a nanoparticle assisted laser desorption/ionization approach that requires low laser energy and has demonstrated high sensitivity. Here we describe NIMS with f-AuNPs for the comprehensive analysis of metabolites in biological tissues. F-AuNPs assist in desorption/ionization by laser-induced release of the fluorocarbon chains with minimal background noise. Since the energy barrier required to release the fluorocarbons from the AuNPs is minimal, the energy of the laser is maintained in the low μJ/pulse range, thus limiting metabolite in-source fragmentation. Electron microscopy analysis of tissue samples after f-AuNP NIMS shows a distinct "raising" of the surface as compared to matrix assisted laser desorption ionization ablation, indicative of a gentle desorption mechanism aiding in the generation of intact molecular ions. Moreover, the use of perfluorohexane to distribute the f-AuNPs on the tissue creates a hydrophobic environment minimizing metabolite solubilization and spatial dislocation. The transfer of the energy from the incident laser to the analytes through the release of the fluorocarbon chains similarly enhances the desorption/ionization of metabolites of different chemical nature, resulting in heterogeneous metabolome coverage. We performed the approach in a comparative study of the colon of mice exposed to three different diets. F-AuNP NIMS allows the direct detection of carbohydrates, lipids, bile acids, sulfur metabolites, amino acids, nucleotide precursors as well as other small molecules of varied biological origins. Ultimately, the diversified molecular coverage obtained provides a broad picture of a tissue's metabolic organization.
Collapse
Affiliation(s)
- Amelia Palermo
- Scripps Center for Metabolomics , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Erica M Forsberg
- Department of Chemistry and Biochemistry , San Diego State University , 5500 Campanile Drive , San Diego , California 92182 , United States
| | - Benedikt Warth
- Department of Food Chemistry and Toxicology, Faculty of Chemistry and Vienna Metabolomics Center (VIME) , University of Vienna , Währingerstraße 38 , 1090 Vienna , Austria
| | - Aries E Aisporna
- Scripps Center for Metabolomics , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Elizabeth Billings
- Scripps Center for Metabolomics , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Ellen Kuang
- Department of Chemistry and Biochemistry , San Diego State University , 5500 Campanile Drive , San Diego , California 92182 , United States
| | - H Paul Benton
- Scripps Center for Metabolomics , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - David Berry
- Department of Microbiology and Ecosystem Science, Division of Microbial Ecology, Research Network Chemistry Meets Microbiology , University of Vienna , Althanstraße 14 , 1090 Vienna , Austria
| | - Gary Siuzdak
- Scripps Center for Metabolomics , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
- Department of Chemistry, Molecular and Computational Biology , The Scripps Research Institute , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| |
Collapse
|
27
|
Singlet Oxygen Generating Properties of Different Sizes of Charged Graphene Quantum Dot Nanoconjugates with a Positively Charged Phthalocyanine. J Fluoresc 2018; 28:827-838. [PMID: 29961202 DOI: 10.1007/s10895-018-2247-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 06/07/2018] [Indexed: 10/28/2022]
Abstract
Various sizes of graphene quantum dots (GQDs) denoted as GQD2, GQD6 and GQD10 (increasing in size) were non-covalently attached to 2,9,16,23-tetrakis[4-(N-methylpyridyloxy)]-phthalocyanine (ZnTPPcQ) to form GQDs-ZnTPPcQ nanoconjugates. X-ray photoelectron spectroscopy (XPS) showed that increasing sizes of GQDs decreases the atomic concentrations of oxygen, which leads to blue shift in spectra of the GQDs. Relative to Pcs alone (0.03), the presence of GQDs improved the singlet oxygen quantum yields with the following values: GQD2-ZnTPPcQ (0.17), GQD6-ZnTPPcQ (0.27) and GQD10-ZnTPPcQ (0.11). GQD2-ZnTPPcQ nanoconjugate system had the most ZnTPPcQ loading, but did not generate the most singlet oxygen species due to aggregation. This study shows that, the quantity of oxygen, size and quality of GQDs as well as amount of Pc loading are amongst the vital properties to consider when constructing GQD-nanoconjugate systems with optimal singlet oxygen quantum yields.
Collapse
|
28
|
Sun S, Huang Y, Zhou C, Chen S, Yu M, Liu J, Zheng J. Effect of Hydrophobicity on Nano-Bio Interactions of Zwitterionic Luminescent Gold Nanoparticles at the Cellular Level. Bioconjug Chem 2018; 29:1841-1846. [DOI: 10.1021/acs.bioconjchem.8b00202] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Shasha Sun
- Department of Chemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Yingyu Huang
- Department of Chemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Chen Zhou
- Department of Chemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Sishan Chen
- Department of Chemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Mengxiao Yu
- Department of Chemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Jinbin Liu
- Department of Chemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Jie Zheng
- Department of Chemistry, The University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
29
|
Nicolardi S, van der Burgt YEM, Codée JDC, Wuhrer M, Hokke CH, Chiodo F. Structural Characterization of Biofunctionalized Gold Nanoparticles by Ultrahigh-Resolution Mass Spectrometry. ACS NANO 2017; 11:8257-8264. [PMID: 28686409 PMCID: PMC5616101 DOI: 10.1021/acsnano.7b03402] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/07/2017] [Indexed: 05/24/2023]
Abstract
Biofunctionalized gold nanoparticles (AuNPs) enable innovative translational research and development in biomedicine. Biomolecules such as peptides, proteins, lipids, and carbohydrates can be assembled onto AuNPs to yield nanomaterials with unique properties for applications in imaging, photothermal therapy, vaccination strategies, and drug delivery. The characterization of functionalized AuNPs still remains an analytical challenge that normally requires the combination of multiple techniques. Laser desorption/ionization (LDI) and matrix-assisted LDI (MALDI) have been applied successfully in combination with time-of-flight (TOF) mass spectrometry (MS) for the analysis of the surface chemistry of AuNPs functionalized with synthetic ligands, however only for ligands with a molecular mass limited to 1000 Da. TOF-MS-based approaches in addition exhibit limited performance in terms of mass resolution and MS/MS possibilities. To overcome these limitations, we designed an approach for the analysis of AuNPs based on ultrahigh resolution Fourier transform ion cyclotron resonance (FTICR) MS and a combination of LDI and MALDI. To illustrate the performance of the method, we present a comprehensive characterization of the surface chemistry of AuNPs conjugated via a thiol-ending linker to either the ovalbumin peptide (OVA 323-339), the Lewis X antigen (Galβ1-4[Fucα1-3]GlcNAcβ1) trisaccharide, the tetramannoside Manα1-2Manα1-2Manα1-3Manα1, or a mixture of both carbohydrates. Collision-induced dissociation (CID) was used to characterize the structure of pseudomolecular ions generated by LDI/MALDI in-depth. These included [M + H]+ and [M + Na]+, and importantly also [M + Au]+ and [M + 2Au-H]+ ions. This first observation of gold-containing pseudomolecular ions provides direct evidence for the Au-conjugation of ligands. In addition, we show the applicability of the method to monitor proteolytic cleavage of peptides that are conjugated to the AuNP surface. The presented LDI/MALDI-FTICR-MS and MS/MS approach will be applicable to the characterization of a wide range of functionalized AuNPs.
Collapse
Affiliation(s)
- Simone Nicolardi
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Yuri E. M. van der Burgt
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Jeroen D. C. Codée
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| | - Manfred Wuhrer
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Cornelis H. Hokke
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
| | - Fabrizio Chiodo
- Center
for Proteomics and Metabolomics and Department of Parasitology, Leiden University Medical Center, Leiden, 2333 ZA, The Netherlands
- Department
of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden University, Leiden, 2333 CC, The Netherlands
| |
Collapse
|
30
|
Ivask A, Mitchell AJ, Malysheva A, Voelcker NH, Lombi E. Methodologies and approaches for the analysis of cell-nanoparticle interactions. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 10:e1486. [DOI: 10.1002/wnan.1486] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/12/2017] [Accepted: 06/20/2017] [Indexed: 01/09/2023]
Affiliation(s)
- Angela Ivask
- Laboratory of Environmental Toxicology; National Institute of Chemical Physics and Biophysics; Tallinn Estonia
- Future Industries Institute; University of South Australia; Mawson Lakes Australia
| | - Andrew J. Mitchell
- Materials Characterisation and Fabrication Platform; University of Melbourne; Melbourne Australia
| | - Anzhela Malysheva
- Future Industries Institute; University of South Australia; Mawson Lakes Australia
| | - Nicolas H. Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences; Monash University; Parkville Australia
| | - Enzo Lombi
- Future Industries Institute; University of South Australia; Mawson Lakes Australia
| |
Collapse
|
31
|
Loutfy SA, Salaheldin TA, Ramadan MA, Farroh KY, Abdallah ZF, Youssef T. Synthesis, Characterization and Cytotoxic Evaluation of Graphene Oxide Nanosheets: In Vitro Liver Cancer Model. Asian Pac J Cancer Prev 2017; 18:955-961. [PMID: 28545193 PMCID: PMC5494245 DOI: 10.22034/apjcp.2017.18.4.955] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: Graphene nanosheets have a broad spectrum of biomedical applications. Hepatocellular cancer (HCC) is a major health problem in the Egyptian population. Currently, treatment strategies are invasive and have several adverse side effects. Thus, other approaches are required for managing this aggressive type of cancer. Our objective here was to prepare and characterize graphene oxide nanosheets and evaluate cytotoxic effect at the molecular level in an in vitro human liver cancer cell model (HepG2). Methods: Graphene oxide nanosheets were generated by chemical oxidation and characterized by transmission electron microscopy and X-ray diffraction. Cytotoxic effects in HepG2 cells were monitored by sulforhodamine B (SRB) colorimetric assay followed by flow cytometric analysis. Molecular investigations of DNA fragmentation and expression of some apoptotic genes at the transcriptional RNA level were also performed. Results: Treatment of HepG2 cells with 400µg/ml graphene oxide nanosheets showed alteration in cell morphology after 24 h. Flow cytometry revealed accumulation of cells in S phase of cell cycle followed by dramatic effects on cellular DNA. Extensive evaluation of the cytotoxic effects of graphene oxide showed increased mRNA Bax apoptotic gene expression with not of P53 and caspase-3 mRNA after 24h, suggesting involvement of an intrinsic apoptotic caspase-independent pathway. Conclusion: Graphene oxide can mediate apoptotic gene signaling in human liver cancer cells opening a novel approach to cancer management. Further analyses at the molecular level are now required to confirm our results and facilitate biomedical applications in vivo.
Collapse
Affiliation(s)
- Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Egypt.
| | | | | | | | | | | |
Collapse
|
32
|
Marsico ALM, Duncan B, Landis RF, Tonga GY, Rotello VM, Vachet RW. Enhanced Laser Desorption/Ionization Mass Spectrometric Detection of Biomolecules Using Gold Nanoparticles, Matrix, and the Coffee Ring Effect. Anal Chem 2017; 89:3009-3014. [PMID: 28193006 DOI: 10.1021/acs.analchem.6b04538] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Nanomaterials have been extensively used as alternate matrices to minimize the low molecular weight interferences observed in typical MALDI but such nanomaterials typically do not improve the spot-to-spot variability that is commonly seen. In this work, we demonstrate that nanoparticles and low matrix concentrations (<2.5 mg/mL) can be used to homogeneously concentrate analytes into a narrow ring by taking advantage of the "coffee ring" effect. Concentration of the samples in this way leads to enhanced signals when compared to conventional MALDI, with higher m/z analytes being enhanced to the greatest extent. Moreover, the ionization suppression often observed in samples with high salt concentrations can be overcome by preparing samples in this way. The ring that is formed is readily visible, allowing the laser to be focused only on spots that contain analyte. The coffee-ring effect represents a new mode by which nanomaterials can be used to enhance the MALDI-based detection of biomolecules.
Collapse
Affiliation(s)
- Alyssa L M Marsico
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - Bradley Duncan
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - Ryan F Landis
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - Gulen Yesilbag Tonga
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| | - Richard W Vachet
- Department of Chemistry, University of Massachusetts , Amherst, Massachusetts 01003, United States
| |
Collapse
|
33
|
Kim YK, Landis RF, Huo S, Kim CS, Vachet RW, Rotello VM. Facile synthesis of cationic gold nanoparticles with controlled size and surface plasmon resonance. RSC Adv 2016; 6:92007-92010. [PMID: 28461886 PMCID: PMC5409524 DOI: 10.1039/c6ra20419h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We present a facile synthetic strategy for large cationic gold nanoparticles by utilizing a cationic thiol ligand as a stabilizer for seed-mediated growth. The size and surface plasmon resonance property of the gold nanoparticles were successfully controlled with this strategy.
Collapse
Affiliation(s)
- Young-Kwan Kim
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- Carbon Convergence Materials Research Center, Institute of Advanced Composite Materials, San 101, Eunha-ri, Bongdong-eup, Wanju-gun, Jeollabuk-do, 565-905 Korea
| | - Ryan F. Landis
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Shuaidong Huo
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
- Carbon Convergence Materials Research Center, Institute of Advanced Composite Materials, San 101, Eunha-ri, Bongdong-eup, Wanju-gun, Jeollabuk-do, 565-905 Korea
| | - Chang Soo Kim
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
34
|
SoRelle ED, Liba O, Campbell JL, Dalal R, Zavaleta CL, de la Zerda A. A hyperspectral method to assay the microphysiological fates of nanomaterials in histological samples. eLife 2016; 5. [PMID: 27536877 PMCID: PMC5042654 DOI: 10.7554/elife.16352] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 08/16/2016] [Indexed: 12/11/2022] Open
Abstract
Nanoparticles are used extensively as biomedical imaging probes and potential therapeutic agents. As new particles are developed and tested in vivo, it is critical to characterize their biodistribution profiles. We demonstrate a new method that uses adaptive algorithms for the analysis of hyperspectral dark-field images to study the interactions between tissues and administered nanoparticles. This non-destructive technique quantitatively identifies particles in ex vivo tissue sections and enables detailed observations of accumulation patterns arising from organ-specific clearance mechanisms, particle size, and the molecular specificity of nanoparticle surface coatings. Unlike nanoparticle uptake studies with electron microscopy, this method is tractable for imaging large fields of view. Adaptive hyperspectral image analysis achieves excellent detection sensitivity and specificity and is capable of identifying single nanoparticles. Using this method, we collected the first data on the sub-organ distribution of several types of gold nanoparticles in mice and observed localization patterns in tumors. DOI:http://dx.doi.org/10.7554/eLife.16352.001 Metallic elements like gold and silver can be made into particles that are one thousand times smaller than the width of a human hair. Researchers can create these “nanoparticles” in different sizes and shapes that exhibit unique properties. For example, gold can be made into rod-shaped particles that interact with infrared light. Other nanoparticles can be loaded with drug molecules and designed to bind to cancer cells. As a result, nanoparticles have been explored for use in a variety of biomedical imaging and therapy applications. However, we must fully understand how the nanoparticles bind to the cancer cells and how the body tolerates these nanoparticles before they can be used in humans. Experiments that explore where nanoparticles accumulate in the body are typically called biodistribution studies. However, current techniques for studying biodistribution cannot simultaneously measure the uptake of particles into organs and reveal the fine structures inside the organs that interact with the particles. SoRelle, Liba et al. aimed to address this problem by developing a new biodistribution technique called HSM-AD (short for hyperspectral microscopy with adaptive detection). This new technique combines a relatively recent method called hyperspectral dark-field microscopy, which can identify nanoparticles from their unique optical signatures, with versatile computer algorithms to detect nanoparticles. HSM-AD is more sensitive than previously developed biodistribution techniques, and SoRelle, Liba et al. used it to produce highly detailed maps of nanoparticle uptake patterns in the organs of mice. These maps provide new insights into how cells and tissues in the body handle different nanoparticles. Moreover, HSM-AD was able to distinguish nanoparticles with unique shapes by their distinct optical signatures. Further experiments show that HSM-AD can reveal interactions between human tumor cells and nanoparticles specifically designed to target those cells. HSM-AD will be a useful resource for researchers studying the effect of nanoparticles on the human body. Future studies will use this technique to explore which nanoparticles have the potential to be developed for medical uses. DOI:http://dx.doi.org/10.7554/eLife.16352.002
Collapse
Affiliation(s)
- Elliott D SoRelle
- Molecular Imaging Program at Stanford, Stanford University, Stanford, United States.,Bio-X Program, Stanford University, Stanford, United States.,Biophysics Program, Stanford University, Stanford, United States.,Department of Structural Biology, Stanford University, Stanford, United States
| | - Orly Liba
- Molecular Imaging Program at Stanford, Stanford University, Stanford, United States.,Bio-X Program, Stanford University, Stanford, United States.,Department of Structural Biology, Stanford University, Stanford, United States.,Department of Electrical Engineering, Stanford University, Stanford, United States
| | - Jos L Campbell
- Molecular Imaging Program at Stanford, Stanford University, Stanford, United States.,Department of Radiology, Stanford University, Stanford, United States
| | - Roopa Dalal
- Department of Ophthalmology, Stanford University, Stanford, United States
| | - Cristina L Zavaleta
- Molecular Imaging Program at Stanford, Stanford University, Stanford, United States.,Department of Radiology, Stanford University, Stanford, United States
| | - Adam de la Zerda
- Molecular Imaging Program at Stanford, Stanford University, Stanford, United States.,Bio-X Program, Stanford University, Stanford, United States.,Biophysics Program, Stanford University, Stanford, United States.,Department of Structural Biology, Stanford University, Stanford, United States.,Department of Electrical Engineering, Stanford University, Stanford, United States
| |
Collapse
|
35
|
Rotello VM. Organic chemistry meets polymers, nanoscience, therapeutics and diagnostics. Beilstein J Org Chem 2016; 12:1638-46. [PMID: 27559417 PMCID: PMC4979691 DOI: 10.3762/bjoc.12.161] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022] Open
Abstract
The atom-by-atom control provided by synthetic organic chemistry presents a means of generating new functional nanomaterials with great precision. Bringing together these two very disparate skill sets is, however, quite uncommon. This autobiographical review provides some insight into how my program evolved, as well as giving some idea of where we are going.
Collapse
Affiliation(s)
- Vincent M Rotello
- Department of Chemistry, University of Massachusetts-Amherst, 710 North Pleasant Street, Amherst, Massachusetts 01003, USA
| |
Collapse
|
36
|
Hou S, Sikora KN, Tang R, Liu Y, Lee YW, Kim ST, Jiang Z, Vachet RW, Rotello VM. Quantitative Differentiation of Cell Surface-Bound and Internalized Cationic Gold Nanoparticles Using Mass Spectrometry. ACS NANO 2016; 10:6731-6. [PMID: 27337000 PMCID: PMC5848210 DOI: 10.1021/acsnano.6b02105] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Differentiation between cell surface-bound and internalized nanoparticles is challenging yet essential for accurately quantifying cellular uptake. Here, we describe a versatile mass spectrometry-based method that allows separate quantification of both cell surface-bound and internalized nanoparticles. This rapid method uses tuned laser fluencies to selectively desorb and ionize cell surface-bound cationic gold nanoparticles from intact cells, providing quantification of external particles. Overall nanoparticle quantities are obtained from the cell lysates, with subtraction of external particles from the total amount providing quantification of taken-up nanoparticles. The utility of this strategy was demonstrated through simultaneous quantitative determination of how cell-surface proteoglycans influence nanoparticle binding and uptake into cells.
Collapse
Affiliation(s)
| | | | - Rui Tang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Yuanchang Liu
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Yi-Wei Lee
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Sung Tae Kim
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Ziwen Jiang
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts Amherst, 710 North Pleasant Street, Amherst, Massachusetts, 01003, USA
| |
Collapse
|
37
|
Aydin F, Dutt M. Surface Reconfiguration of Binary Lipid Vesicles via Electrostatically Induced Nanoparticle Adsorption. J Phys Chem B 2016; 120:6646-56. [DOI: 10.1021/acs.jpcb.6b02334] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Fikret Aydin
- Department
of Chemical and
Biochemical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Meenakshi Dutt
- Department
of Chemical and
Biochemical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
38
|
Loutfy SA, Al-Ansary NA, Abdel-Ghani NT, Hamed AR, Mohamed MB, Craik JD, Eldin TAS, Abdellah AM, Hussein Y, Hasanin MTM, Elbehairi SEI. Anti-proliferative Activities of Metallic Nanoparticles in an in Vitro Breast Cancer Model. Asian Pac J Cancer Prev 2016; 16:6039-46. [PMID: 26320493 DOI: 10.7314/apjcp.2015.16.14.6039] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AIMS To investigate effect of metallic nanoparticles, silver (AgNPs) and gold nanoparticles (AuNPs) as antitumor treatment in vitro against human breast cancer cells (MCF-7) and their associated mechanisms. This could provide new class of engineered nanoparticles with desired physicochemical properties and may present newer approaches for therapeutic modalities to breast cancer in women. MATERIALS AND METHODS A human breast cancer cell line (MCF-7) was used as a model of cells. Metallic nanoparticles were characterized using UV-visible spectra and transmission electron microscopy (TEM). Cytotoxic effects of metallic nanoparticles on MCF-7 cells were followed by colorimetric SRB cell viability assays, microscopy, and cellular uptake. Nature of cell death was further investigated by DNA analysis and flow cytometry. RESULTS Treatment of MCF-7 with different concentrations of 5-10nm diameter of AgNPs inhibited cell viability in a dose-dependent manner, with IC50 value of 6.28μM, whereas treatment of MCF-7 with different concentrations of 13-15nm diameter of AuNPs inhibited cell viability in a dose-dependent manner, with IC50 value of 14.48μM. Treatment of cells with a IC50 concentration of AgNPs generated progressive accumulation of cells in the S phase of the cell cycle and prevented entry into the M phase. The treatment of cells with IC50 concentrations of AuNPs similarly generated progressive accumulation of cells in sub-G1 and S phase, and inhibited the entrance of cells into the M phase of the cell cycle. DNA fragmentation, as demonstrated by electrophoresis, indicated induction of apoptosis. CONCLUSIONS Our engineered silver nanoparticles effectively inhibit the proliferation of human breast carcinoma cell line MCF-7 in vitro at high concentration (1000 μM) through apoptotic mechanisms, and may be a beneficial agent against human carcinoma but further detailed study is still needed.
Collapse
Affiliation(s)
- Samah A Loutfy
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt E-mail :
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Du R, Zhu L, Gan J, Wang Y, Qiao L, Liu B. Ultrasensitive Detection of Low-Abundance Protein Biomarkers by Mass Spectrometry Signal Amplification Assay. Anal Chem 2016; 88:6767-72. [PMID: 27253396 DOI: 10.1021/acs.analchem.6b01063] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A mass spectrometry signal amplification method is developed for the ultrasensitive and selective detection of low-abundance protein biomarkers by utilizing tag molecules on gold nanoparticles (AuNPs). EpCAM and thrombin as model targets are captured by specific aptamers immobilized on the AuNPs. With laser desorption/ionization time-of-flight mass spectrometry (LDI-TOF MS), the mass tag molecules are detected to represent the protein biomarkers. Benefiting from the MS signal amplification, the assay can achieve a limit of detection of 100 aM. The method is further applied to detect thrombin in fetal bovine serum and EpCAM in cell lysates to demonstrate its selectivity and feasibility in complex biological samples. With the high sensitivity and specificity, the protocol shows great promise for providing a new route to single-cell analysis and early disease diagnosis.
Collapse
Affiliation(s)
- Ruijun Du
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and Institutes of Biomedical Sciences, Fudan University , Handan Road 220, Shanghai 200433, China
| | - Lina Zhu
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and Institutes of Biomedical Sciences, Fudan University , Handan Road 220, Shanghai 200433, China
| | - Jinrui Gan
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and Institutes of Biomedical Sciences, Fudan University , Handan Road 220, Shanghai 200433, China
| | - Yuning Wang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and Institutes of Biomedical Sciences, Fudan University , Handan Road 220, Shanghai 200433, China
| | - Liang Qiao
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and Institutes of Biomedical Sciences, Fudan University , Handan Road 220, Shanghai 200433, China.,Shanghai Stomatological Hospital, Fudan University , East Beijing Road 356, Shanghai 200001, China
| | - Baohong Liu
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers and Institutes of Biomedical Sciences, Fudan University , Handan Road 220, Shanghai 200433, China.,Shanghai Stomatological Hospital, Fudan University , East Beijing Road 356, Shanghai 200001, China
| |
Collapse
|
40
|
Abdelhamid HN, Wu HF. Gold nanoparticles assisted laser desorption/ionization mass spectrometry and applications: from simple molecules to intact cells. Anal Bioanal Chem 2016; 408:4485-502. [DOI: 10.1007/s00216-016-9374-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 11/29/2015] [Accepted: 01/28/2016] [Indexed: 01/05/2023]
|
41
|
Affiliation(s)
- Ian L. Gunsolus
- Department of Chemistry, University of Minnesota, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| | - Christy L. Haynes
- Department of Chemistry, University of Minnesota, 207 Pleasant
Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
42
|
Marsico ALM, Elci GS, Moyano DF, Yesilbag Tonga G, Duncan B, Landis RF, Rotello VM, Vachet RW. Enhanced Laser Desorption/Ionization Mass Spectrometric Detection of Gold Nanoparticles in Biological Samples Using the Synergy between Added Matrix and the Gold Core. Anal Chem 2015; 87:12145-50. [DOI: 10.1021/acs.analchem.5b02985] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Alyssa L. M. Marsico
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Gokhan S. Elci
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Daniel F. Moyano
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Gulen Yesilbag Tonga
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Bradley Duncan
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Ryan F. Landis
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Richard W. Vachet
- Department of Chemistry, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
43
|
Ulusoy M, Jonczyk R, Walter JG, Springer S, Lavrentieva A, Stahl F, Green M, Scheper T. Aqueous Synthesis of PEGylated Quantum Dots with Increased Colloidal Stability and Reduced Cytotoxicity. Bioconjug Chem 2015; 27:414-26. [DOI: 10.1021/acs.bioconjchem.5b00491] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | | | | | | | | | | | - Mark Green
- Department
of Physics, King’s College London, The Strand, WC2R 2LS London, U.K
| | | |
Collapse
|
44
|
Marsico ALM, Creran B, Duncan B, Elci SG, Jiang Y, Onasch TB, Wormhoudt J, Rotello VM, Vachet RW. Inkjet-printed gold nanoparticle surfaces for the detection of low molecular weight biomolecules by laser desorption/ionization mass spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2015; 26:1931-1937. [PMID: 26202457 DOI: 10.1007/s13361-015-1223-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 06/22/2015] [Accepted: 06/26/2015] [Indexed: 05/24/2023]
Abstract
Effective detection of low molecular weight compounds in matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS) is often hindered by matrix interferences in the low m/z region of the mass spectrum. Here, we show that monolayer-protected gold nanoparticles (AuNPs) can serve as alternate matrices for the very sensitive detection of low molecular weight compounds such as amino acids. Amino acids can be detected at low fmol levels with minimal interferences by properly choosing the AuNP deposition method, density, size, and monolayer surface chemistry. By inkjet-printing AuNPs at various densities, we find that AuNP clusters are essential for obtaining the greatest sensitivity. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Alyssa L M Marsico
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Brian Creran
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Bradley Duncan
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - S Gokhan Elci
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Ying Jiang
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | | | | | - Vincent M Rotello
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA
| | - Richard W Vachet
- Department of Chemistry, University of Massachusetts-Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
45
|
Bai Y, Xing H, Wu P, Feng X, Hwang K, Lee JM, Phang XY, Lu Y, Zimmerman SC. Chemical Control over Cellular Uptake of Organic Nanoparticles by Fine Tuning Surface Functional Groups. ACS NANO 2015; 9:10227-36. [PMID: 26327513 DOI: 10.1021/acsnano.5b03909] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The functional groups displayed on the surface of nanoparticles (NP) are known to play an important role in NP cellular uptake. However, only a few systematic studies have been reported to address their role, in large part because of the difficulty in regularly varying the number and structure of the functional groups on the NP surface. We employ a bottom-up strategy for the synthesis of water-soluble organic nanoparticles (ONPs) with different sizes and functional groups, using readily available monomers. Utilizing flow cytometry, we measured the HeLa cell uptake efficiency of ONPs that contain side-chains with a different (a) length, (b) number of hydroxyl groups, and (c) number of methyl groups. We have also investigated ONPs with the same functional groups but different sizes. The potential formation and influence of protein corona was examined using the same approach but in the presence of serum. The results demonstrate that under both serum and serum-free conditions the surface-exposed functional groups determine the efficiency of cellular uptake of the particles, and that the trend can be partially predicted by the lipophilicity of the polymeric ONP's repeating units. Also, by using a "masking" strategy, these particles' cellular uptake behavior could be altered conveniently.
Collapse
Affiliation(s)
- Yugang Bai
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Hang Xing
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
- Beckman Institute, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Peiwen Wu
- Department of Biochemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Xinxin Feng
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Kevin Hwang
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Jennifer M Lee
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Xin Yi Phang
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Yi Lu
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
- Beckman Institute, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
- Department of Biochemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | - Steven C Zimmerman
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| |
Collapse
|
46
|
Timerbaev AR. Role of mass spectrometry in the development and medicinal implementation of metal-based nanoparticles. JOURNAL OF ANALYTICAL CHEMISTRY 2015. [DOI: 10.1134/s1061934815090166] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
47
|
Supramolecular nanoscale assemblies for cancer diagnosis and therapy. J Control Release 2015; 213:152-167. [PMID: 26160308 DOI: 10.1016/j.jconrel.2015.06.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/26/2015] [Accepted: 06/27/2015] [Indexed: 02/08/2023]
Abstract
Nanocarriers based on polymers, metals and lipids have been extensively developed for cancer therapy and diagnosis due to their ability to enhance drug accumulation in cancer cells and decrease undesired drug toxicity in healthy tissues. Overcoming multidrug resistance by designing proper drug nanocarriers will improve outcome of existing oncologic treatments such as chemotherapy and radiotherapy. In this article the relation between physicochemical properties and capacity of a nanosystem to deliver therapeutic agents into pathological sites is discussed. Most promising examples of drug delivery systems are reviewed, and, in particular, the design of a carbohydrate based matrix with entrapped gold nanoparticles is highlighted.
Collapse
|
48
|
García I, Henriksen-Lacey M, Sánchez-Iglesias A, Grzelczak M, Penadés S, Liz-Marzán LM. Residual CTAB Ligands as Mass Spectrometry Labels to Monitor Cellular Uptake of Au Nanorods. J Phys Chem Lett 2015; 6:2003-2008. [PMID: 26266492 DOI: 10.1021/acs.jpclett.5b00816] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Gold nanorods have numerous applications in biomedical research, including diagnostics, bioimaging, and photothermal therapy. Even though surfactant removal and surface conjugation with antifouling molecules such as polyethylene glycol (PEG) are required to minimize nonspecific protein binding and cell uptake, the reliable characterization of these processes remains challenging. We propose here the use of laser desorption/ionization mass spectrometry (LDI-MS) to study the ligand exchange efficiency of cetyltrimethylammonium bromide (CTAB)-coated nanorods with different PEG grafting densities and to characterize nanorod internalization in cells. Application of LDI-MS analysis shows that residual CTAB consistently remains adsorbed on PEG-capped Au nanorods. Interestingly, such residual CTAB can be exploited as a mass barcode to discern the presence of nanorods in complex fluids and in vitro cellular systems, even at very low concentrations.
Collapse
Affiliation(s)
- Isabel García
- †CIC biomaGUNE, Paseo de Miramón 182, 20009 Donostia-San Sebastián, Spain
- ‡Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Paseo de Miramón 182, 20009 Donostia-San Sebastián, Spain
| | | | | | - Marek Grzelczak
- †CIC biomaGUNE, Paseo de Miramón 182, 20009 Donostia-San Sebastián, Spain
- §Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Soledad Penadés
- †CIC biomaGUNE, Paseo de Miramón 182, 20009 Donostia-San Sebastián, Spain
- ‡Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Paseo de Miramón 182, 20009 Donostia-San Sebastián, Spain
| | - Luis M Liz-Marzán
- †CIC biomaGUNE, Paseo de Miramón 182, 20009 Donostia-San Sebastián, Spain
- ‡Biomedical Research Networking Center in Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Paseo de Miramón 182, 20009 Donostia-San Sebastián, Spain
- §Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
49
|
Bhardwaj V, Srinivasan S, McGoron AJ. Efficient intracellular delivery and improved biocompatibility of colloidal silver nanoparticles towards intracellular SERS immuno-sensing. Analyst 2015; 140:3929-34. [PMID: 25939798 DOI: 10.1039/c5an00435g] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
High throughput intracellular delivery strategies, electroporation, passive and TATHA2 facilitated diffusion of colloidal silver nanoparticles (AgNPs) are investigated for cellular toxicity and uptake using state-of-art analytical techniques. The TATHA2 facilitated approach efficiently delivered high payload with no toxicity, pre-requisites for intracellular applications of plasmonic metal nanoparticles (PMNPs) in sensing and therapeutics.
Collapse
Affiliation(s)
- Vinay Bhardwaj
- 10555 West Flagler Street, Department of Biomedical Engineering, Florida International University, Miami, FL, USA.
| | | | | |
Collapse
|
50
|
Kim C, Tonga GY, Yan B, Kim CS, Kim ST, Park MH, Zhu Z, Duncan B, Creran B, Rotello VM. Regulating exocytosis of nanoparticles via host-guest chemistry. Org Biomol Chem 2015; 13:2474-2479. [PMID: 25569869 PMCID: PMC4323993 DOI: 10.1039/c4ob02433h] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Prolonged retention of internalized nanoparticulate systems inside cells improves their efficacy in imaging, drug delivery, and theranostic applications. Especially, regulating exocytosis of the nanoparticles is a key factor in the fabrication of effective nanocarriers for chemotherapeutic treatments but orthogonal control of exocytosis in the cellular environment is a major challenge. Herein, we present the first example of regulating exocytosis of gold nanoparticles (AuNPs), a model drug carrier, by using a simple host-guest supramolecular system. AuNPs featuring quaternary amine head groups were internalized into the cells through endocytosis. Subsequent in situ treatment of a complementary cucurbit[7]uril (CB[7]) to the amine head groups resulted in the AuNP-CB[7] complexation inside cells, rendering particle assembly. This complexation induced larger particle assemblies that remained sequestered in the endosomes, inhibiting exocytosis of the particles without any observed cytotoxicity.
Collapse
Affiliation(s)
- Chaekyu Kim
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Gulen Yesilbag Tonga
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Bo Yan
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Chang Soo Kim
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Sung Tae Kim
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Myoung-Hwan Park
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Zhengjiang Zhu
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Bradley Duncan
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Brian Creran
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| | - Vincent M. Rotello
- Department of Chemistry, University of Massachusetts, 710 North Pleasant Street, Amherst, MA 01003, USA
| |
Collapse
|