Novel Nanomaterials for Protein Analysis

Novel Nanomaterials for Protein Analysis

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS Pengyuan Yang*, Liming Wei*,† Key Laboratory of Medical Epigenetics and Metabolism, Institutes o...

2MB Sizes 49 Downloads 199 Views

CHAPTER

2

NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

Pengyuan Yang*, Liming Wei*,† Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences & Department of Chemistry & The Fifth People’s Hospital of Shanghai, Fudan University, Shanghai, China* Shanghai Songjiang District Central Hospital, Shanghai, China†

INTRODUCTION As the product of gene transcription in biology, protein has become an important role due to its pertinence to functional genomics. The analysis of protein is focused on the identification and quantification to unravel protein structures, functions, localizations, interactions, and posttranslational modifications in the complicated samples. With the development of novel nanomaterials, a category of powerful platforms was established for protein analysis. Here, we will summarize recent progresses in the application of novel nanomaterials for protein analysis.

PROTEIN ANALYSIS As the central cellular components in biological networks, proteins play an important role in diverse biological functions including cytoskeletal building blocks, biochemical reactions, enzymatic catalysis, immune response, or transcription factors affecting gene expression [1–5]. Protein has become a key factor to explore the biological sciences and a source of potential biomarkers and drug targets [6–8]. Therefore, protein analysis at a system level has come into existence to narrow the existing gaps between discovery sciences and the associated clinical applications. Proteomics has attracted considerable interest in discerning the global biological processes. Originally put forwarded in 1995 by Marc Wilkins, proteomics is the science that explores the dynamic of proteins in response to physiological, pathological, and environmental influences [9]. The first dimension structure of protein is its amino acid sequence, providing a link between proteins and their coding genes via the genetic code [10, 11]. The divergence of protein expression level in different phenotypes might support the discovery of important regulator factors, potential biomarkers, biological mechanisms, and even drug targets. Moreover, the spatial partition of proteins at a subcellular level provides information regarding protein function, interactions, and cellular mechanisms [12]. Furthermore, as an essential cellular regulatory mechanism, posttranslational modifications (PTMs) can alter the activities of protein targets [13], and the cumulative effect of PTMs in biology can regulate the large signaling pathways and networks in cell. Finally, the activities and functions of proteins are further modulated by Novel Nanomaterials for Biomedical, Environmental and Energy Applications. https://doi.org/10.1016/B978-0-12-814497-8.00002-3 Copyright # 2019 Elsevier Inc. All rights reserved.

37

38

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

interacting proteins in metabolic and signaling pathways [14]. Consequently, proteomics aims to characterizing the whole proteome profile and further explore the biological mechanism and clinical applications.

CHALLENGES IN PROTEIN ANALYSIS With the rapidly growing field of protein analysis, proteomics has embedded in several fields in biology, including disease pathogenesis, biomarker discovery, and pharmacology of drugs. However, the analysis of whole proteins in cell or biology species presents several challenges. First, the large-scale protein analysis of proteome is more complicated. Due to the alternative splicing, mutation, and integral chemical modifications of mRNA used for protein expression, highly complex proteins are expressed in biology samples [15, 16]. As a result, it is estimated that a single gene can generate up to 10–20 proteoforms [17]. Additionally, there are approximately 430 PTMs that contribute to the complexity of proteins [18]. Second, the high dynamic range difference of expressed proteins hampers the detection of low-abundance protein. Due to responses to activation, turnover, downregulation, conformation, or localization, proteins are expressed in different abundance in biological samples. In a blood sample, more than 10,000 different proteins are expressed with a dynamic range of nine orders of magnitude. Consequently, proteins at concentrations of 1012–1015 M usually coexist with those at concentrations of 103–104 M, making detection of low-abundance proteins extremely difficult [19]. Meanwhile, compared with gene analysis with tools such as polymerase chain reaction (PCR) for the amplification of a single nucleic acid molecule in a complex biological sample up to the limit of detection by modern instruments, detection of ultralow-abundance proteins remains a great challenge with the absence of such tool in protein analysis. Therefore, development of ultrasensitive, robust, and high-throughput analytic techniques that can rapidly detect proteins in biological sample is in urgent need in the protein analysis.

NANOMATERIALS FOR PROTEIN ANALYSIS Various nanomaterials can be categorized into carbon-based or silicon-based nanomaterials, metallic nanomaterials, metal oxide nanomaterials, semiconductor nanomaterials, and polymer nanomaterials according to its texture [20]. The application of nanomaterials in biology has been of great interest in recent years due to the chemically tunable sizes and unique physical (e.g., electronic, photonic, and magnetic) properties and has been applied in various biological frontier including imaging [21], biosensing [22], target drug/gene delivery [23], biological conjugates, and biomacromolecule surface recognition [24]. A novel technical platform known as nanoproteomics utilizes the advantages of nanotechnology to help tackle those aforementioned challenges presented in protein analysis such as protein complexity and dynamic range. Due to large surface-to-volume ratio and chemical functional group on the surface, nanomaterials as sorbent materials have been extensively applied in the sample pretreatment of proteins, such as pretreatment of low-abundance proteins or peptides, specific posttranslational modification of proteins or peptides, and immobilization of enzyme to increase the efficiency of digestion. Meanwhile, the electric and optical properties of nanomaterials can also support it as matrix substrate or detection probe for enhancing protein detection sensitivity. Furthermore, nanomaterials also place the important role in the protein quantification as labeling probe. In summary, the unique physical and chemical properties

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

39

of nanomaterials, such as high surface-to-volume ratios, variable surface functionality, and adjustable size or structure, offer several advantages in nanoproteomics including low sample consumption, high-throughput capability, short assay time, and sensitive detection. Herein, we summarize the recent nanomaterials that are applied in various protein analyses.

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS In a living organism, tens or thousands of proteins are expressed in a dynamic range of ten orders of magnitude. Meanwhile, there are hundreds of modifications decorated on proteins posttranslationally for different biological functions. Thus, to promote the efficiency for protein identification and characterization, more efficient sample pretreatment methods were developed using novel nanomaterials. Here, various novel nanomaterials are reviewed in the sample preparation of protein analysis.

PRETREATMENT OF LOW ABUNDANCE PROTEINS OR PEPTIDES Mass spectrometry-based proteomics analysis using soft ionization techniques such as matrix-assisted laser desorption/ionization (MALDI) and electrospray ionization (ESI) has become a major player in protein profiling search due to capabilities of these techniques to perform protein analysis with high throughput and excellent sensitivity [25, 26]. However, the identification of low-abundance proteins has encountered some challenges including the inevitable sample loss and coconcentration with the contaminants in protein analysis. Thus, an efficient pretreatment step prior to MS analysis can aid identification of low-abundance proteins. Nanomaterials have been employed for pretreatments of low-abundance proteins or peptides. Among the diverse classes of nanomaterials, silica-based nanomaterials [27, 28], carbon-based nanomaterials [29, 30], and polymer nanomaterials are the most commonly used composites that offer several advantages in pretreatment of low-abundance proteins or peptides, including low sample consumption, ultralow detection, and short assay time (Fig. 2.1).

CNTs, chitosan/MWCNTs, MWCNTs@Fe3O4-MIPs, etc.

Fe3O4@mSiO2 Fe3O4@nSiO2@mSiO2 Fe3O4@nSiO2@mSiO2-C8 etc.

C8-modified GO@mSiO2, G@SiO2@PMMA, GO/rGO-Fe3O4, Fe3O4@SiO2@GO, etc.

Fe3O4@SiO2-C8, Fe3O4-NH2C8,Fe3O4@C-C8, etc.

NDs

HSBF, C60-f-MSs SBA-15

MCM-41

Silica-based

Carbon-based nanomaterials

ZnO(TiO2)@PMMA, Fe3O4@SiO2@PMMA, polymer brush nanosponge, etc.

Polymer-based

FIG. 2.1 Schematic illustration of low-abundance proteins or peptide enrichment for MS detection using diverse nanomaterials.

40

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

Silica-based nanomaterials Silica has remarkable properties such as large specific surface area, hydrophilic and hydrophobic surface function, making it a promising material for the enrichment of trace proteins or peptides. Nanozeolite beta (b-X, X ¼ Si/Al ratio) and silicalite-1 (S-1) nanocrystals were applied for the first time in the enrichment and identification of trace peptides and proteins [27]. Nanozeolites with a medium or high Si/Al ratio exhibited best features on the enrichment of tryptic myoglobin digests with no apparent discrimination. Furthermore, with the high specific surface areas and ordered porous structures with narrow size distribution, mesoporous materials have opened up new applications to adsorb or capture large biomolecules through the interaction with the functional groups of pore and surface. As wellordered hexagonal mesoporous silicate molecular sieves, SBA-15 were used for the capturing proteins and also as a multifunctional nanoreactor where digests of proteins can be done. After the evaluation of the protein adsorption isotherms for standard proteins, SBA-15 exhibited efficient adsorption. Meanwhile, due to the digestion process was reacted inside the channels, sample loss was decreased compared with normal procedure [28]. Furthermore, a novel mesoporous silica particles with critical ˚ can perform selective extraction of pore sizes with a narrow pore size distribution centering at 20.5 A endogenous peptides (from 1 to 12 kDa) from human plasma and exclusion of untargeted proteins at the same time [31]. Considering the pH conditions of endogenous peptides, highly ordered mesoporous nanoparticles MCM-41 were modified with strong cation-exchange and anion-exchange groups to the enrichment of endogenous peptides at natural pH condition [32]. Then, combined enrichment with three kinds of materials (SCX-MCM-41, SAX-MCM-41, and unmodified MCM-41) and online 2-D nano-LC/MS/MS analysis, a large-scale identification of endogenous peptides from mouse liver extract was reported. To enhance the enrichment efficiency, the multifunctional magnetic materials combining with mesoporous silica and iron oxides were fabricated with highly ordered pores, special surface features, and superparamagnetic properties [33, 34]. For instance, the novel microspheres Fe3O4@nSiO2@mSiO2 composed of the Fe3O4@nSiO2 core and a perpendicularly aligned mesoporous SiO2 shell were prepared, and a 100-fold increase of S/N ratio of standard peptides was observed. To directly prepare a mesoporous SiO2 shell on the magnetic microspheres (Fe3O4@mSiO2 spheres), nonporous silica composites were dissolved in the alkaline solution with the template cetyltrimethylammonium bromide (CTAB), and upon completion, the template interacted with silicate species was removed by ammonium nitrate ethanol solution [34].

Carbon-based nanomaterials Carbon-based nanomaterials have many promising applications due to their unique properties such as good chemical stability and high specific surface area. Additionally, this surface modification of this type of material can be easily achieved and has been extensively studied. Consequently, carbon-based nanomaterials have also been widely functionalized as novel adsorbents for sample pretreatment in protein analysis. The adsorption mechanism relies on the affinity between the proteins or peptides, and carbon nanomaterials stem from hydrophobic interactions and sometimes π-stacking interactions between the materials and aromatic residues such as histidine and tryptophan. Carbon nanomaterials, such as carbon nanotubes, fullerenes (buckyballs), graphene, and nanodiamond, can be applied for the pretreatment of low-abundance proteins or peptides from biological samples. In order to improve the specific enrichment and dispersity of carbon nanomaterials in aqueous solution, some modification or functionalization of carbon nanomaterials has been fabricated and employed to improve their performance.

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

41

The carbon nanotubes (CNTs), including single-walled nanotubes (SWNTs) and multiwalled nanotubes (MWNTs), could be readily modified via covalent or noncovalent functionalization. Chen et al. pretreated CNTs with citric acid to obtain CNTs with anionic surfaces, which could be used to selectively concentrated proteins or peptides with opposite charge by varying the pH of the sample solution [29]. Meanwhile, with the assistance of its electronic properties, peptides adsorbed on CNTs can be analyzed by MALDI-MS without the need of any extra MALDI matrix [30]. Guo et al. derivatized the surface of CNTs using 2,5-dihydroxybenzoyl hydrazine, which introduced phenolic hydroxyl and phenyl groups [35]. Combined mechanical and electronic properties of CNTs and a source of protons by the derivatized molecules, the functionalized CNTs could be placed as assisted matrix for the MALDI-MS analysis, on which the detection could be enhanced about 10–100 times. Meanwhile, 2521 endogenous peptides have successfully been identified from human plasma assisted by CNTs. For the specific isolation of hemoglobins, a chitosan/MWCNT composite has been fabricated by cross-link. The novel composite exhibited good enrichment properties than conventional chitosan particles with micrometer-range dimension including faster adsorption equilibrium and higher adsorption capacity [36]. Furthermore, to enhance the enrichment specificity, a novel magnetic MWCNTs coated with a layer of molecularly imprinted polymers (MWCNTs@Fe3O4-MIPs) were synthesized for specific enrichment of some template proteins [37]. C60 fullerene and its derivatives have become more popular in biological applications due to their unique properties including well-defined structure and strong absorption in the UV region. A star-like water-soluble fullerene derivative, hexa(sulfonbutyl)fullerene (C60[(CH2)4SO3]6, HSBF), was used for the enrichment of positively charged species through ion interaction and hydrophobic interaction [38]. With strong absorbance in the UV range, proteins or peptides enriched using fullerene derivatives can be deposited onto a target as a mixture and analyzed by MALDI-MS. Fullerene (C60)-functionalized silica gel has been fabricated for the pretreatment of peptides and other biomolecules [39, 40]. Further, polymerization of C60 on the surfaces of Fe3O4@SiO2 magnetic nanoparticles (MNPs) (designated C60-f-MS) was synthesized and successfully applied for the enrichment of low-concentration tryptic cytochrome C with good salt tolerance capacity (100 mM CaCl2)[41]. As some commercial C8/C18-functionalized SiO2 beads have been applied for the desalting of proteins or peptides, the alkyl group-functionalized MNPs were designed for the pretreatment of proteins/peptides in biological samples through hydrophobic interaction [42–44]. In the initial study, C8 groups were successfully immobilized on the surface of magnetic silica microsphere with the routine silanization using chloro(dimethyl)octylsilane [42]. In the following work, alkyl groups were simply anchored on the outside of amine-functionalized MNPs (Fe3O4-NH2). To improve the dispersibility and magnetic response, C8 group was loaded on magnetic carbonaceous polysaccharide (Fe3O4@C) MNPs. Due to the hydrophilic groups of carbonaceous polysaccharide bonded outside the spheres, the hydrophilicity and stability of the novel Fe3O4@C-C8 spheres were improved in aqueous systems. These well-dispersed Fe3O4@C-C8 spheres also have a strong magnetic response, in which the spheres can be collected within 2 s [44]. Owing to the excellent stability, biocompatibility, and unbleachable fluorescing from point defects, nanodiamond (ND) has emerged as a new material for applications in nanoproteomics [45, 46]. Chang et al. successfully utilized the carboxylated/oxidized-NDs for protein enrichment [47]. Notably, since diamonds are optically transparent up to the UV region, the enriched proteins or peptides can be directly analyzed by MALDI-MS without eluted from NDs. Yang et al. established the on-plate enrichment procedure to peptides by detonation NDs (dNDs), and the detection sensitivity using these

42

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

materials was enhanced by more than two orders of magnitude. Meanwhile, dNDs could be used for the enrichment of peptides in highly diluted and complex sample solutions. As dNDs were prepared in extreme environment during detonation process, dNDs have a variety of surface functional groups. As a result, enrichment using this material can be achieved in a wide pH range [48]. As the new generation of carbon materials, graphene (G) is a single carbon atom thick and twodimensional honeycomb crystal lattice, and this material has recently attracted much attention in biological application [49]. Owing to an ultrahigh specific surface area (theoretical value 2630 m2 g1) and the large delocalized p-electron system, graphene has a strong affinity to biomolecule with carbonbased ring structures. G and GO were firstly fixed on different silica NPs through covalent bonding or electrostatic interactions [50–52]. Graphene-bound silica exhibited good desalting of protein samples for MALDI-TOF MS analysis with good enrichment features (recoveries of protein ranged from 82.4% to 94.7%). To simplify the enrichment process, Fe3O4 particles were covalently attached onto the GO sheets. The prepared Fe3O4-GO composites can be applied for immobilization and pretreatment of low-abundance proteins or peptides that have abundant hydrophilic and hydrophobic groups with high loading capacity [53]. It’s noteworthy that Fe3O4@SiO2@graphene microspheres with perfect core-shell structure were fabricated and could be reusable for protein enrichment [54]. Recently, to improve the hydrophilicity of magnetic-functionalized graphene, Zhang et al. established a Fe3O4chitosan@graphene (Fe3O4-CS@G) for the pretreatment of low-abundance proteins [55].

Polymer nanomaterials With the development of biopharmaceuticals, the composite of polymers and nanoparticles has been widely used as drug delivery vehicles for protection and slow release. Due to the flexible properties and activated functional groups, polymer nanomaterials have drawn attention on sample preparation for protein or peptides analysis. As poly(methyl methacrylate) (PMMA) possesses a large number of hydrophilic and hydrophobic groups, it has potential promise for fabricated a series of PMMA-coated nanoparticles for the pretreatment of proteins or peptides. Yang and coworkers prepared CaCO3@PMMA with a destructible CaCO3 core, which can be dissolved with acetic acid after pretreatment and desalination of proteins or peptides. The proteins or peptides enriched on PMMA were released into solution for MALDI-MS analysis [56]. Moreover, PMMA shell was polymerized on the ZnO and TiO2 NPs by virtue of the inherent free radicals for the pretreatment of proteins or peptides [57–59]. This synthesis method could make stable ZnO and TiO2 cores, and the resulting PMMA nanobeads are dispersive in solution. Due to the small diameters of the ZnO@PMMA NPs and TiO2@PMMA NPs, enrichment experiments using these nanoparticles could be directly spotted on the MALDI plate without an eluting step. Good enrichment features (limit of detection (LOD) of tryptic bovine serum albumin (BSA) peptides, 100 aM) and high salt tolerance (1 nM L1 BSA tryptic peptides in NaCl (6.2 M), saturated NH4HCO3 (2.6 M), or 1 M urea aqueous solution) were observed. PMMA was functionalized on Fe3O4@SiO2 MNPs with the assistance of 3methacryloxypropyltrimethoxysilane (MPS), a polymerizable silane coupling agent [60]. On gold substrate, a novel polymer brush-like nanosponge was developed by 70% poly(N-isopropylacrylamide) (PNIPAAM) and 30% poly(methacrylic acid) (PMAA), in which the structure could be adjusted with different pH condition and realized the separation and enrichment of peptides/proteins [61]. In a later study, a cationic polymer brush nanosponge was also reported for the selective extraction and enrichment of acidic proteins and peptides [62].

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

43

PRETREATMENT OF PHOSPHORYLATED PROTEINS OR PEPTIDES Protein phosphorylation is a vital PTM that dynamically modulates numerous biological processes controlled by the protein kinases and phosphatases [63, 64]. Phosphorylation places important roles in many biological processes, including the cell signaling networks and communication, gene expression, protein synthesis, and cell division and apoptosis [65, 66]. Thus, phosphorylated proteins or peptides are very important for illuminating the specific signaling pathways and finding new targets for the diagnosis and therapy of diseases. MS-based proteomics is a powerful tool for the identification of phosphorylated proteins or peptides. However, due to the low abundance and worse ionization efficiency of phosphorylated proteins or peptides in MS analysis compared with their unphosphorylated parts, sample pretreatment of phosphorylated proteins or peptides prior to MS is necessary for their effective detection. Based on ionic interaction, metal ion-immobilized nanoparticles and nanostructure metal oxides were fabricated and applied for the specific pretreatment of phosphorylated proteins or peptides. Nanomaterial-based metal ion-immobilized particles focus on the application of new metal ions, the introduction of new chelating ligands, and the fabrication of novel nanomaterial substrates. Different novel nanomaterials for the pretreatment of phosphorylated proteins or peptides are summarized as follows.

Metal ion-immobilized composites Generally, the phosphate groups show typical Lewis-base properties in a wide pH range. Thus, the phosphate groups on the phosphorylated proteins/peptides can interact with metal cations by electrostatic interaction or/and chelation. New metal ions, new chelating ligands, and novel nanomaterial substrates have been developed for the immobilized metal ion affinity chromatography (IMAC) techniques in the phosphorylated protein analysis (Table 2.1).

Metal ions Metal ions including Al3+, Cu2+, Fe3+, Zn2+, Ti4+, and Zr4+ have been utilized in IMAC strategies (Table 2.1). As the trivalent/tetravalent ions of the stable rare earth can strongly interact with phosphate group, rare earth metal materials have attracted interest by researchers in the biomedical frontiers several years ago. Therefore, rare earth ions such as Ga3+ and Ce4+ have also been used as immobilized metal ion in IMAC strategy for the phosphoprotein or phosphopeptide isolation. With iminodiacetic acid (IDA) as a chelating ligand, Ce4+ was immobilized onto magnetic silica microspheres (Fe3O4@SiO2) and shown better enrichment capacity than the Fe3O4@SiO2-IDA-Fe (III) [81]. Further, Ni et al. coated poly(vinylphosphonic acid) (PVPA) as a chelating ligand for the immobilization of Ce(IV) onto the Fe3O4@SiO2. As Ce4+ has unique properties such as good dispensability in solutions and high abundance after coating, the Fe3O4@SiO2@PVPA-Ce(IV) showed good sensitivity and specificity for the enrichment of phosphorylated peptides from biological samples [82]. Moreover, other rare earth ions including La3+, Ho3+, and Er3+ have been fixed on PVPA spheres for the pretreatment of phosphorylated proteins [83].

Chelating ligand and matrix supports The traditional ligands including IDA and nitrilotriacetic acid (NTA) was applied for immobilizing metal ions (e.g., Fe3+ and Ga3+) [67, 68, 70]. The immobilized metal ions with only one IDA ligand may be easily lost during sample loading and washing. As the chelating ligand, NTA was much better

44

Table 2.1 Nanomaterials for Pretreatment of Phosphorylated Proteins or Peptides Based on IMAC

Fe (III) Al (III) Ga (III) Ti(IV)

Zr (IV)

Zn(II)

Ce (IV)

Type of Matrix Fe3O4@SiO2-IDA-M

Chelating Ligand

Characterization of Matrix

Selectivity (BSA/α/ β-Casin)

Sensitivity

References

50:1



[67]

GLYMO-IDA NTA, IDA

d ¼ 370 nm Ms ¼ 68.2 emu g1 d ¼ 15 nm –

– –

5 fmol –

[68] [69]

Fe3O4@SiO2-EDAS-SANTA-Mn+ MCNC@PMAA@PEG MP Fe3O4-ATP-Mn+

EDAS-SA-NTA

d ¼ 50 nm



[70]

Fe3O4@PD-Mn+

Dopamine

500:1

SiO2@graphene

1000:1

Fe3O4@SiO2-PO3-Mn+

Dihydroimidazole group –PO3-OH

d ¼ 70 nm

100:1

Fe3O4@C-PO3-Mn+

–PO3-OH

d ¼ 300 nm

25:1

Fe3O4@p(VPA-EDMA-1) Mn+

–PO3-OH

50:1

Fe3O4@SiO2-AsO3-Mn+

Arsenate group

Fe3O4-GAPT-Mn+

Glutaric acid

d ¼ 400–600 nm (20 nm of polymer shell) Ms ¼ 29 emu g1 d ¼ 30 nm Ms ¼ 39.1 emu g1 d ¼ 4–6 nm

50 fmol tryptic digest of α/β-casein 0.5 nM tryptic digest of β-casein 3 amol tryptic digest of β-casein 5 fmol tryptic digest of β-casein 40 pM tryptic digest of β-casein 50 fmol tryptic digest of β-casein 0.8 pmol tryptic digest of β-casein –

CoFe2O4-GAPT-Mn+ Fe3O4@SiO2-IDA-Mn+

Glutaric acid GLYMO-IDA

Fe3O4@SiO2@PVPA-Mn+

–PO3-OH

n+

GLYMO-IDA

Fe3O4@SiO2-IDA-Mn+ Ni-NTA/IDA-Mn+

500:1 ATP

d ¼ 150 nm

d ¼ 9.96  1.03 nm d ¼ 370 nm Ms ¼ 68.2 emu g1 d ¼ 500 nm Ms ¼ 50.5 emu g1

5000:1

100:1

10 fmol tryptic digest of β-casein

99:1 (mass ratio)

50:1

[71] [72] [73] [74] [75] [76] [77]

[78] [79]

1.15 μM β-casein

[80] [81]

5 nM β-casein

[82]

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

Metal ion

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

45

than IDA due to the optimized enrichment efficiency with four or six coordinate number. Meanwhile, 2 some novel chelating ligands including arsenate (–AsO2 3 ), phosphate (–PO3 ), adenosine triphosphate (ATP), and dopamine were developed to enhance the coordination force [78, 84]. Moreover, to further increase the coordination capacity and simplify the pretreatment process, various silica-based, carbonbased, polymer-based, and magnetic-based matrix supports have been utilized to incorporate these ligands. To enhance the stability and specificity for the enrichment of multiply phosphorylated peptides, an easy and cost-effective method to prepared modified arsenate groups (As) on silica-based and magnetic silica nanoparticle matrix materials was developed, respectively [78, 84] (Fig. 2.2A). The zirconium arsenate-modified magnetic nanoparticles (ZrAs-Fe3O4@SiO2) and ZrAs-silica nanoparticles show many potential applications in clinical research. Meanwhile, based on an octahedral coordination model with phosphate groups, phosphate ligands for metal ion immobilization were established on porous silicon wafer and silica nanoparticles by the chemical reaction between –NH2 and POCl [75, 85] (Fig. 2.2B). To improve the performance of phosphate-group ligand-functionalized matrix, with the assisted polymeric matrices, Fe3O4@SiO2@PEG-Ti4+ affinity microspheres were developed obtained through polymerization of monomers carrying reactive groups (e.g., epoxy and amino) (Fig. 2.2C) [86]. Fe3O4@SiO2@PEG-Ti4+ from previous discussion has the following benefits including simple isolation during experimental steps due to its magnetic core, minimal nonspecific protein adsorption due to PEG chains, and high abundance of Ti4+ on the surface. Hence, other polymer-related ligands were developed for the immobilization of metal ion with good selectivity, high recovery, and an ultralow detection limit, such as MCNC@PMAA@p(EGMP)-Ti4+ composite and Fe3O4@SiO2@ (HA/CS)10-Ti4+ [71, 87]. Other novel ligands also place important roles for the improvement of the specificity, sensitivity, and enrichment capacity for phosphorylation [72]. With the help of adenosine triphosphate (ATP) molecules, Ti4+ was grafted onto Fe3O4 nanoparticles with high sensitivity (3 amol β-casein). Due to easily polymerized onto a variety of substrate, dopamine was used for the immobilization of Ti4+ ions through the strong chelation. The prepared Fe3O4@ polydopamine (PD)-Ti4+ exhibited good specificity (molar ratio of tryptic BSA/β-casein up to 500:1) and high sensitivity (2 fmol tryptic β-casein) [73]. Additionally, the unique hydrophilicity of dopamine also allows the resulting nanoparticles to have excellent water dispersibility and reduced nonspecific binding. Another exceptional novel affinity ligand is dipicolylamine (DPA), which formed Zn-DPA coordination complex on the surface of superparamagnetic NPs (4.6  0.7 nm) via glutaric acid (hereafter referred to as GAPT) [79], as illustrated in Fig. 2.2D. Attributed to the super small size, the reduced nonspecific adsorption due to the PEG chains, and the high abundance of Zn2+, the prepared Fe3O4-GAPT-Zn NPs (4–6 nm) gave outstanding specificity (mass ratio of BSA/β-casein up to 99:1). Significantly, the Fe3O4-GAPT-Zn NPs could perform specific enrichment of phosphorylated proteins in human plasma and showed good compatibility with the top-down proteomics technique for the analysis of phosphorylated protein from a swine heart tissue extract. Recently, the author designed and synthesized the cobalt ferrite (CoFe2O4) NPs (9.96  1.03 nm) as the core instead of Fe3O4 for the preparation of the CoFe2O4-GAPT-Zn NPs, which showed a stronger magnetic response and better reproducibility for intact phosphorylated protein enrichment than the first generation Fe3O4-GAPT-Zn NPs [80].

46

Representative works in the pretreatment of phosphorylated proteins/peptides. (A) Zirconium arsenate-modified magnetic nanoparticles (ZrAsFe3O4@SiO2) (Copyright 2011, Springer-Verlag), (B) ZrP-functionalized mesoporous silica microspheres (Copyright #2011, Royal Society of Chemistry), (C) Fe3O4@SiO2@PEG-Ti4+ affinity microspheres microspheres (Copyright 2012, Royal Society of Chemistry), and (D) functionalized Fe3O4-GAPT-Zn NPs (Copyright 2015, American Chemical Society).

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

FIG. 2.2

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

47

Metal oxide based nanomaterials Metal oxide (MO) nanoparticles or core/shell nanocomposites are promising materials for the selective phosphorylation enrichment, which also used metal ions as affinity sites to phosphorylated proteins/ peptides. Different with IMAC materials, in which metal ions was chelated with ligand on the surface of matrix, the metal ions from metal oxide-based nanomaterials bind to their neighbor oxygen ions via strong chemical bonds. Some metal oxides are amphoteric, for example, TiO2 with pKa1 ¼ 4.4 and pKa2 ¼ 7.7, which can capture phosphorylation proteins/peptides at low pH and then be eluted at high pH based on the reversible Lewis acid-base actions [88].

Pure metal oxide nanomaterials Most of pure metal oxides in their common form have been used for the specific enrichment of phosphorylated proteins or peptides including TiO2 [89], ZrO2 [90], CeO2 [91], NiO [92], Fe2O3 [93], HfO2 [94], Al2O3 [95], Ga2O3 [96], ZnO [97], SnO2 [98], La2O3 [99], and Ta2O3 [100]. In an evaluation test, the authors observed that ZrO2 has greater specificity to single phosphorylated peptides, while TiO2 favored multiply phosphorylated peptides [90]. CeO2 and Ce-Zr composite, which exhibited dual properties in phosphopeptide enrichment, work as a sorbent for phosphorylated peptides and a catalyst for dephosphorylation [101]. In MS spectrum, the signal of phosphorylated peptides treated with CeO2 showed a characteristic mass loss of 80 Da, which may be useful for the direct identification of phosphorylated peptides. Other metal nanomaterials including Fe3O4, NiFe2O4, ZnFe2O4, and NiZnFe2O4 have shown distinct specificity to mono- and multiply phosphorylated peptides [102]. Recently, with the unique properties including high surface area, stable skeleton, and size exclusion effect (with pore sizes typically between 2 and 50 nm), mesoporous metal oxides showed an even higher efficiency and loading capacity for binding phosphate groups than micro- and nanoparticles [91, 103]. For example, with a high binding capacity due to its surface area, mesoporous TiO2 has been used as a special adsorbent for phosphorylated peptides. It has the advantage of both the specific affinity with TiO2 and the size exclusion mechanism enabled by the mesoporous structure. Meanwhile, with the highly hydrophilic outer surface of each cluster, nonspecific binding of many hydrophobic proteins or peptides can be mitigated [104]. Further, superparamagnetic iron oxide core allows the material-target conjugate isolation from the solution using an external magnet after selective adsorption [105].

Magnetic core based MOs Magnetic core-based MOs usually have core-shell structures. A variety of metal oxides (TiO2 [106], ZrO2 [107], Al2O3 [95], ZnO [79], Ta2O5 [108], Nb2O5 [109], Ga2O3 [96], SnO2 [110], and CeO2 [91]) can be functionalized on the outside of magnetic cores. Deng et al. developed a series of synthetic procedure for the preparation of metal oxide-coated MNPs (Fe3O4@MxOy) with a well-defined core-shell structure [95, 100, 106, 107, 110]. A solvothermal and polymerization reaction was applied for the preparation of Fe3O4 microspheres and the carbon layer, respectively. Finally, metal oxide was prehydrolyzed and adsorbed onto the Fe3O4@C microspheres, by a sol-gel process and calcination. TiO2, ZrO2, Al2O3, and Ga2O3 were successfully coated on Fe3O4 MNPs using this procedure [110]. To prepare new metal oxide-coated MNPs with high surface area and porous structure, either sol-gel method or liquid-phase deposition method was applied for the immobilization of metal oxide on magnetic hollow mesoporous silica sphere for the enrichment of phosphorylated peptides [91]. Wang et al. coated a highly crystalline mesoporous TiO2 shell via sol-gel method followed by a hydrothermal process [104].

48

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

The mesoporous Fe3O4@mTiO2 microspheres possess a well-defined core/shell structure, large pore volume (0.45 cm3 g1), high specific surface area (167.1 m2 g1), and high magnetic susceptibility. Due to the purity and high density of the TiO2 mesoporous shells, the Fe3O4@mTiO2 microspheres showed over 10 times as that of the Fe3O4@TiO2 microspheres (20 mg g1). Considering the improvement of the specificity and sensitivity and the mitigation of “shadow effect”, Ni et al. fabricated the Fe3O4@fTiO2 multifunctional microspheres with flowerlike hierarchical meso-/macroporous TiO2 shells [111]. Compared with the former materials with macroporous shell (denoted as Fe3O4@pTiO2), Fe3O4@fTiO2 exhibited excellent specificity and selectivity for the enrichment of phosphorylated peptides. With chemical stability, catalysis, and coordination, some rare earth (RE) composites have also been immobilized on the outside of magnetic core as novel affinity absorbents for the pretreatment of phosphorylated peptides. The Fe3O4@REPO4 microspheres (RE ¼ La and Y) were synthesized via precipitation and ion-exchange method [112, 113]. 2678 unique phosphorylated peptides had been identified by Fe3O4@hYPO4 microspheres from the tryptic digested mouse brain. Furthermore, other RE-based materials were also applied for the enrichment and detection of phosphorylated peptides, such as magnetic Fe3O4@LaxSiyO5, γ-Fe2O3@REVO4 (RE ¼ Sm, Dy, and Ho), and γ-Fe2O3@xNH4FyLuF3 [114–116].

Other nanomaterials-based MOs To increase the dispersibility of MOs in aqueous solution, carbon-based nanomaterials have been utilized as matrices for supporting MOs. Recently, TiO2/CNTs composites and TiO2/graphene composites are the most attractive materials of choice [117, 118]. With assisted by graphene, a graphene-titania platform (G-TiO2) was established by sol-gel method and applied for detecting phosphorylated peptides in HeLa cells. Notably, with the UV absorbance of graphene and titania, G-TiO2 can be utilized as a substrate for the direct MS analysis of phosphorylated peptides and simplified experimental workflow [119]. To improve affinity sites, G@PD@TiO2 composite was fabricated by coating PD on graphene for the deposition of TiO2 nanoparticles [120]. With the assistance of PD, the G@PD@TiO2 possesses a large amount of TiO2 and good dispersibility and showed good enrichment features, with which 334 phosphopeptides can be identified from mouse brain tissue. With its unique properties, the modifiable surface of silica is an ideal feature for the fabrication of MO-based nanomaterials. For instance, with large surface area and appropriate Lewis acidity of the TiO2- and ZrO2-modified mesoporous silica composites, they exhibited better enrichment performance than commercial available TiO2 particles [103, 121]. Furthermore, to overcome the “shadow effect” in some mesoporous materials with long channels, the macroporous silica with large and open pores has also been applied for the MOs coating to facilitate efficient mass transport. The Al2O3 and TiO2 onto the macroporous ordered silica foam (MOSF) were fabricated and used as a multifunctional reactor with enzyme immobilization and the substrate for the subsequently pretreatment of phosphorylated peptides [122]. Subsequently, Deng et al. designed the synthesis of TiO2-modified hierarchically ordered macro-/mesoporous silica composites [123]. Besides metal ion-immobilized nanoparticles and MO-modified nanomaterials, other nanomaterials have been successfully fabricated for the specific enrichment of phosphorylated proteins/peptides. For instance, amine-based nanomaterials are expected to capture phosphorylated proteins/peptides by the ionic interaction controlled by the pH of buffers. With the various styles of amine groups, some molecules have been successfully functionalized on the surface of nanomaterials (Table 2.2).

Type

Chelating Molecules

Fe3O4@NH2 nanoparticles

1,6-Hexane-diamine

C60fullerene@aminopropylsilica Fe3O4@SiO2-NH2

Amino-propylsilica

Characterization of Matrix

Selectivity (BSA/α/ β-Casin)

Sensitivity

References

d ¼ 370 nm Ms ¼ 68.2 emu g1 d ¼ 3–5 μm

50:1



[124]



2.2 μM β-casein

[125]

d ¼ 15 nm



5 fmol

[126]

Fe3O4@SiO2@PEI Diamond nanoparticles@PA

N0 -[3-(trimethoxysilyl)propyl]-diethylenetriamine and tetramethylorthosilicate Polyethylenimine (PEI) Polyarginine(PA)

– d ¼ 50 nm

– –

[127] [128]

Graphene@guanidyl

1,6-Hexanediamine



500:1

Fe3O4@PGMA

Poly(glycidyl methacrylate) (PGMA)

d ¼ 150 nm

5000:1

Fe3O4@SiO2@PGMAguanidyl

3-Guanidopropyl triethoxysilane

d ¼ 360 nm

500:1

– 50 fmol tryptic digest of α/βcasein 0.5 nM tryptic digest of β-casein 3 amol tryptic digest of β-casein 5 fmol tryptic digest of β-casein

[129]

[130]

[131]

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

Table 2.2 Nanomaterials for Pretreatment of Phosphorylated Proteins/Peptides Based on Amine Group

49

50

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

The typical 1,6-hexanediamine-functionalized magnetic nanoparticles (Fe3O4@NH2) can capture phosphorylated peptides in the 1% TFA buffer, and the peptides can be eluted in the eluting buffer containing 5% NH3H2O [124]. To increase the abundance of amino group on the composite surface, some polymerized amino molecules were coated on the surface of magnetic particles by sol-gel method and polymerization reaction. Fe3O4@SiO2@ polyethylenimine (PEI) carrying abundant amine groups can capture phosphorylated peptides by forming bidentate complexes [127]. Through stable “covalentlike” interaction with phosphate groups, guanidine-related nanomaterials was synthesized and applied for the enrichment of phosphorylated peptides for direct MALDI-MS analysis. With the affinity mechanism similar to the MOs, the magnetic metal-organic frameworks (mMOFs) have also been synthesized via electrostatic self-assembly between positively charged MIL-101(Fe) and the negatively charged Fe3O4 magnetic nanoparticles (MNPs) [132]. With highly specific surface area, excellent magnetic property, and well-defined structures, Fe3O4/MIL-101(Fe) composite exhibited good specificity, recovery, and application performance for real samples.

PRETREATMENT OF GLYCOPROTEINS/GLYCOPEPTIDES Protein glycosylation plays important factors in various cellular processes, such as gene expression, cell adhesion, signal transduction, and immune responses. Due to its close related with disease biomarkers, glycosylation contains much information that has been extensively studied using proteomics tools. For example, some glycosylated proteins were recognized by FDA as cancer biomarkers, including human chorionic gonadotropin-β, α-fetoprotein, carcinoembryonic antigen, prostate-specific antigen, human epidermal growth factor receptor 2, and mucins, which are clinically applied in diagnosis, monitoring, or selected therapies [133]. In glycosylation protein analysis, the complex process requires the identification of the peptide sequence and the glycosylation site and the characterization of glycan composition and the structure. The routine strategy of the glycosylated protein analysis is to perform enzymatic digestion followed by glycosylated peptide sequencing through mass spectrometry, which has been widely applied in glycoproteomics research. However, due to the low abundant and low ionization efficiency of glycosylated proteins/peptides and the microheterogeneity of each glycosylation site, rapid and effective pretreatment strategies before MS analysis are urgently needed for glycoproteomics. Currently, some effective pretreatments of glycosylated proteins/peptides are realized based on three aspects: (a) hydrophilic interaction, (b) chemical reaction, and (c) lectin affinity. Recently, with the growing attention on glycosylation, various functionalized nanomaterials based on the above mentioned principles have been synthesized for the pretreatment of glycosylated proteins/peptides.

Hydrophilic-interaction based pretreatment There are various materials that utilize the principles of hydrophilic interaction, such as siloxane, zwitterionic (ZIC)-based, amide-based, and carbohydrate-modified materials. Based on the hydrophilicity of the glycan moiety, some hydrophilic group-modified nanomaterials have been developed for the pretreatment of glycosylated proteins or peptides with the advantages including good reproducibility, high selectivity, and nonirreversible alterations of glycosylated proteins/peptides (Table 2.3). Meanwhile, hydrophilic interaction-based method utilizes high composition of organic solvent as reaction buffer, making the workflow compatible with MS analysis. Glycosylated proteins or peptides contain abundant hydroxyl and carboxylic acid groups, especially in negatively charged sialylated and sulfated glycosylated peptides, which may interact with positive ions via electrostatic interaction. For instance,

51

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

Table 2.3 Nanomaterials for Pretreatment of Glycosylated Proteins/Peptides Based on HILIC Interaction Type Silica Fe3O4@ NH2

Functional Moiety Silica hydroxyl Amine

Phenolic mesoporous polymers

Amine

Amine-functionalized mesoporous silica nanoparticles (SBA-15APTES) MIL-101(Cr)NH2@PAMAM Magnetic particlecarbohydrate conjugates Fe3O4@SiO2@PEGmaltose

Amine

Highly cross-linked chitosan microspheres (CSMs) PAM-g-allosex@SiO2 Cross-linked CD-MOFs (LCD-MOFs) GO-Fe3O4/SiO2/ AuNWs/L-Cys magG/PDA/Au/L-Cys

Amine

Characterization of Support

Real Sample

References

d ¼ 1.7 μm

CD44 fusion protein Mouse uterine luminal fluid Human serum

[134]

[136]

Human serum

[137]

– MP-70000-220, BET surface area (548 m2 g1), mesopore size (13 nm) The content of –NH2 in the SBA-15-NH2 was 45.55  105 mol g1 Surface area, 778.11 m2 g1

Carbohydrate

Carbohydrate

Carbohydrate

Size, Fe3O4 core (240 nm); silica shell (6 nm); PEG shell (20 nm); Ms, 35.9 emu g1 Surface area and pore size, 191 m2 g1 and 10 nm

Carbohydrate



γ-Cyclodextrin

Cubic particle size, 200–300 nm –

Poly(MBAAm-coMAA)@L-Cys MIL-101(NH2)@AuCys Water-soluble zwitterionic Au NCs Histidine bonded silica (HBS) Carbonized Mil-101 (Cr)

Amino acid (cysteine) Amino acid (cysteine) Amino acid (cysteine) Amino acid (cysteine) Amino acid (cysteine) Amino acid (histidine) Size-exclusion effect

SiO2-RAFT@PMSA

Zwitterionic

[135]

[138] Human sera

[139]



[140]

Rat brain

[141]

HeLa S3 cell lysate Mouse liver

[142] [143] [144]

Au nanoparticles was 60 nm

Mouse liver proteins Human serum

d ¼ 200 nm

Human plasma

[146]

Surface areas, 240.4 m2 g1

HeLa cell lysate Mouse liver

[147]

Average diameter 2.0 nm The surface coverage of histidine was 0.9 μmol m2 BET surface area of 531.88 m2 g1; pore diameter, 3.03 nm

Human serum digest Human serum

Mouse liver glycoproteome

[145]

[148] [149] [150]

[151]

Continued

52

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

Table 2.3 Nanomaterials for Pretreatment of Glycosylated Proteins/Peptides Based on HILIC Interaction—cont’d Type

Functional Moiety

Fe3O4@SiO2@PMSA

Zwitterionic

TCP-SMs Fe3O4@PDA@ZrSO3H CS@PGMA@IDATi4+ Titanium dioxide

Zwitterionic Zwitterionic

Characterization of Support

Real Sample

References

Mouse liver

[152]

Mouse liver Human serum

[153] [154]

Ti4+

Size, Fe3O4 core 280 nm; silica shell 10 nm, PMSA layer 60 nm d ¼ 50100 nm Ms 26.4 emu g1; pore size, 3.2 nm d ¼ 20250 nm

Mouse liver

[155]

TiO2

d ¼ 5 μm

[156]

MCNC@PMAA@AgNPs

Ag+

[157]

Graphitic carbon nitride (g-C3N4)-

Triazine ring and N–H



[158]

Magnetic graphene functionalized with metal-organic frameworks (MOFs) (MG@Zn-MOFs)

Triazine ring and N–H

Size, magnetic colloid nanocrystal cluster (MCNC) core 308 nm; PMAA shell 36 nm; Ag NP shell 10–30 nm; Ms, 12.1 emu g1 Surface area of 10.6 m2 g1 and a pore volume of 0.03 m3 g1 Size, Fe3O4 core 200 nm; ZIF-8 crystals 30 nm (BET) surface area 322 m2 g1, pore ˚ 10.6 A

HeLa cell membranes Rat serum

Human serum

[159]

the bare silica phase with silanol surface groups is a type of hydrophilic material, which can concentrate the glycan chain of glycosylated proteins or peptides in the hydration layer around the silica particles via electrostatic forces and hydrogen bonding [134]. Based on the hydrophilic interaction, some novel nanomaterials have been fabricated for the selective enrichment of glycosylated proteins or peptides from complex sample.

Amide group based hydrophilic interaction Generally, the amide stationary phase is currently the most frequently used HILIC material for separating glycans and glycopeptides, as shown in a recent survey by Zauner et al. [160]. Therefore, the amide group was applied for the preparation of absorbent to glycosylated proteins/peptides. For example, with the hydrophilic and ionic interaction between amino group and glycan structure, aminefunctionalized Fe3O4 MNPs could be directly applied for the enrichment of glycosylated peptides, especially on the sialic acid/sulfate residue of glycosylated peptides [135]. Khoo et al. prepared and used amine-functionalized Fe3O4 MNPs for the identification of glycosylated peptides from mouse uterine luminal fluid [135]. The percentage of glycoprotein identification increased from 70% using Sepharose CL-4B to 77% using NH2-Fe3O4 MNPs. From the obtained data, NH2-functionalized

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

53

nanoparticles performed favorably against the Sepharose CL-4B enrichment method selected for direct comparative evaluation and, in fact, against any other nonselective glycopeptide enrichment. Dai et al. proposed a new template-free strategy for the fabrication of phenolic mesoporous polymers (MPs) for highly selective glycosylated peptide enrichment (Fig. 2.3A). The resulting MPs showed a number of attractive features, including high surface areas (613 m2 g1), large accessible porosities (pore volume, 0.53 cm3 g1), nitrogen-containing functionalities, and intrinsic hydrophilic skeletons, that facilitate highly specific and efficient enrichment with excellent sensitivity of glycosylated peptides from minute

FIG. 2.3 Different nanomaterials applied in the pretreatment of glycosylated proteins/peptides. (A) Phenolic mesoporous polymers (MPs) (Copyright 2015, American Chemical Society), (B) Fe3O4@SiO2@PEG-maltose MNPs (Copyright 2012, Royal Society of Chemistry), (C) PAM-g-allosex@SiO2 (Copyright 2016, American Chemical Society), (D) Fe3O4@SiO2@PMSA (Copyright 2014, Royal Society of Chemistry), (E) Fe3O4@PDA@Zr-SO3H (Copyright 2017, Elsevier B.V.), (F) poly(MBAAm-co-MAA)@L-Cys (Copyright 2016, American Chemical Society), (G) MIL-101(NH2)@Au-Cys (Copyright 2017, American Chemical Society), (H) Fe3O4@L-Cys (Copyright 2017, Springer-Verlag GmbH Germany), (I) MCNC@PMAA@Ag-NPs (Copyright 2012, Royal Society of Chemistry), (J) zirconia-coated mesoporous silica (ZrO2/MPS) (Copyright 2012, Royal Society of Chemistry), (K) magnetic graphene functionalized with metal-organic frameworks (MOFs) (MG@Zn-MOFs) (Copyright 2016, American Chemical Society), and (L) graphitic carbon nitride (g-C3N4) (Copyright 2017, American Chemical Society).

54

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

amounts of biological samples, where they identified 169 unique glycopeptides and 92 glycoproteins in 5 μL human serum. Thus, it opens up new opportunities for the development of glycosylated peptides enrichment [136]. Further, Gao et al. synthesized an amino-functionalized metal-organic framework (MOF) MIL-101(Cr)-NH2 whose surface is grafted with a hydrophilic dendrimer poly(amidoamine) (PAMAM) for N-glycopeptide enrichment based on hydrophilic interaction [138]. The selected substrate MOF MIL-101(Cr) possesses high surface area that provides support for peptide adsorption. In addition, the MOF displayed a good hydrophilic property after being modified with amino groups. The MIL-101(Cr)-NH2 material showed to have low detection limit (1 nm standard glycoprotein horseradish peroxidase digestion) and good selectivity when the concentration of nonglycosylated peptides was 100-fold higher than the target N-glycopeptides.

Carbohydrate-based ligands Carbohydrate-based ligands have emerged as specialized hydrophilic interaction materials, and these carbohydrates include monosaccharides, polysaccharides [139], cellulose, chitosan [141], and cyclodextrins in the stationary phase. These carbohydrates are often linked on the stationary phase via click chemistry, which is a chemical approach to quickly and reliably join small precursors for a desired conjugated moiety (Fig. 2.3B and C). Zou et al. fabricated hybrid Fe3O4@SiO2@PEG-Maltose MNPs to achieve large binding capacity by conjugation on the PEG molecules (Fig. 2.3B) [140]. This strategy utilizes the terminal hydroxyl group of the PEG molecule, which can be chemically modified to azido functionality for further functionalization via click chemistry. The existence of PEG molecule provides enhanced water solubility for the nanoparticles in aqueous solutions. The availability of high abundance of hydroxyl groups on maltose groups can form complex hydrogen-bonding networks for cooperative binding with the oligosaccharide of glycosylated proteins/peptides via multivalent hydrophilic interactions. As a result, enhancement of the sensitivity and higher sequence coverage for glycosylated proteins/peptides were achieved. To enrich trace sialylated glycosylated peptides from highly complex samples, a novel sialylated receptor, allose units were integrated into a polyacrylamide chain and generated a saccharide-responsive smart copolymer (SRSC) according to the saccharide-saccharide interactions in life systems [142]. This design significantly improves the selectivity of sialylated acid binding (500-fold BSA) and exhibits high-performance enrichment capacity. In Hela S3 cell lysate, more than 200 sialylated glycosylated peptides with 180 unique sialylated glycosylation sites were successfully characterized from 50 μg protein samples pretreated by PAM-g-allosex@SiO2 in two replicated experiments (Fig. 2.3C).

Zwitterionic molecules functionalized nanomaterials Zwitterionic molecule-functionalized materials are well-known super-hydrophilic and environmental materials. Generally, zwitterionic groups on nanoparticles can bind water molecules more strongly via electrostatically induced hydration, in comparison with water retention materials based on hydrogenbonding induced hydration. Thus, zwitterionic groups enable the formation of a layer of water molecules, which greatly contributes to hydrophilic interaction. Recently, additional hydrophilic interaction novel nanomaterials were prepared for glycosylated peptides enrichment, such as the zwitterionic polymer and “thiol-ene” cysteine (TE-Cys) bound to different supports (Fig. 2.3D and E). The thick zwitterionic polymer shell grants the nanomaterials not only with excellent hydrophilic surface properties but also with abundant zwitterionic functional sites on the polymer chains for high detection sensitivity (0.1 fmol), large binding capacity for glycosylated peptides (100 mg g1), and high enrichment

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

55

efficiency (above 73.6%). As a promising zwitterionic monomer, cysteine can contain both positive and negative charges simultaneously at suitable pH and is capable of enhancing the efficiency of glycosylated peptides enrichment via hydrophilic interaction (Fig. 2.3F–H). Cysteine was functionalized on the surface of different nanomaterials via Au or other functional group [145, 147, 161]. Zhang et al. synthesized a novel zwitterionic-hydrophilic material named poly (N,N-methylenebisacrylamidecomethacrylic acid)@L-Cys (poly(MBAAm-co-MAA)@L-Cys), via the combination of distillation precipitation polymerization and “thiol-ene” click reaction (Fig. 2.3F) [146]. To attain high enrichment sensitivity and capacity for the pretreatment of glycosylated proteins/peptides, a novel ultrathin Au nanowire-assisted zwitterionic-hydrophilic magnetic graphene oxide (GO-Fe3O4/SiO2/AuNWs/L-Cys) was synthesized with combined properties of individual components including the good biocompatibility of GO, strong magnetic responses of Fe3O4, large surface area of ultrathin Au nanowires, and finally excellent hydrophilicity of L-Cys via four simple and rapid synthetic steps [144]. Due to its one-dimensional structure, the ultrathin Au nanowires were easily grafted with an abundant amount of L-Cys, which attributed to the excellent enrichment performance for glycosylated proteins/peptides, including low detection limit (10 fmol of human Ig G and 26 glycosylated peptides) and large binding capacity (150 mg g1). In addition to conventional hydrophilic interaction, several other variations for this technique have also recently emerged, such as metal-based hydrophilic nanomaterials or carbon-based nanomaterials, for the enrichment of glycosylated proteins/peptides (Fig. 2.3I–L). Owing to the multivalent interaction between glycosylated peptides and silver nanoparticles (Ag-NPs), an Ag-NP functional shell was coated on the outside of a magnetic core via a PMAA interim layer (Fig. 2.3I) [157]. Meanwhile, TiO2 NPs can also be applied for the specific enrichment of sialic acid (SA)-containing glycosylated peptides via multipoint binding, and a protocol for the comprehensive analysis of SA-containing glycosylated peptides was established. In a combination of filter-aided sample preparation (FASP), TiO2 enrichment, multiple enzyme digestion, and high pH reversed-phase peptide separation, an optimized strategy was used for the establishment of SA glycosylation database in human plasma, in which a total of 982 glycosylation sites were identified in 413 proteins [156]. Furthermore, a shell of zeolitic imidazolate framework coated on magnetic graphene (MG@Zn-MOFs) (Fig. 2.3K) and a novel graphitic carbon nitride (g-C3N4) were also prepared and applied for the pretreatment of low-abundance glycosylated proteins/peptides and SA-containing glycosylated peptides (Fig. 2.3L) [158, 159].

Chemistry based pretreatment As the cis-diol group on the carbohydrate is present in glycan structures, pretreatment strategy was developed for the enrichment of glycosylated proteins or peptides without discrimination among different glycan structures. The chemical reactions involved in glycosylated proteins/peptides pretreatment include boronic acid chemistry, hydrazide chemistry, reductive amination chemistry, oxime click chemistry, coordination chemistry, and alkyne click chemistry. Lu et al. has given the detailed review to their respective mechanism and applications [162]. In the following section, the nanomaterials developed for the boronic acid chemistry and hydrazide chemistry method were discussed and summarized.

Boronic acid chemistry based nanomaterials Boronic acid can be reacted with the cis-diol group on the sugar moieties by producing five- or sixmembered cyclic esters in nonaqueous or basic aqueous media [163]. Importantly, this covalent cyclic ester is reversible in an acidic pH, and therefore, this mechanistic feature has been exploited to isolate

56

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

glycosylated proteins/peptides. Since boronic acid is stable, readily available, and their reactivity has been extensively studied, incorporation of these molecules onto solid supports is relatively simple, especially nanomaterials. Magnetic nanoparticles (MNPs) have become an ideal support for the functionalization of boronic acid groups for pretreatment of glycosylated proteins/peptides before MS analysis as the resulting nanomaterials can be easily isolated via an external magnet. Aminophenylboronic acid (APBA)functionalized MNPs (Fe3O4@SiO2-CHO-APBA) were synthesized through multistep covalent modification [164]. To simplify the multistep chemical procedure, the same authors first prepared the amine-functionalized MNPs (Fe3O4-NH2), followed by synthesis of the phenylboronic acidfunctionalized disperse core-shell MNPs (Fe3O4@SiO2@VPBA) using polymerization reaction and sol-gel method [165]. To further improve the abundance of boronic acid functionalized on MNPs, Au or polymer was also used to synthesize core-shell structure including Fe3O4@C@Au [166], Fe3O4@SiO2@Au [167], dendrimeric boronic acid-functionalized MNPs [168], and Fe3O4@P (AAPBA-co-monomer) NPs [169]. Due to the large amount of the functionalization group of the above nanomaterial, the boronic acid-functionalized magnetic nanoparticles exhibited significantly enhanced binding strength for glycosylated proteins. For example, the dendrimeric boronic acid-functionalized MNPs gave the dissociation constants of 105–106 M for glycoproteins, which was three to four orders of magnitude higher than the affinity associated with single boronic acid binding [168]. Besides MNPs, various novel nanomaterials have also been applied as solid supports in boronic acid chemistry-based approaches, and these nanomaterials exhibited remarkable performance for the pretreatment of glycosylated proteins/peptides owing to their unique nanostructures. For instance, boronic acid-functionalized FDU-12 (FDU-12-GA) and MCM-41 were synthesized for the enrichment of glycosylated peptides [170, 171]. The detection limit of glycopeptides had two orders of magnitude improvement by the prepared boronic acid-functionalized FDU-12 (FDU-12-GA) compared with traditional analysis. With the combination of the size exclusion effect of MCM-41 and the selectivity of boronic acid chemistry, this material was used in glycopeptidome research with excellent selectivity (glycopeptide/nonglycopeptide analyses at molar ratios of 1:100), ultralow sensitivity (femtomolelevel detection limitation), good binding capacity (40 mg g1), and high recovery of glycopeptides (up to 88.10%). With the assisted alkyl linker, aminophenylboronic acid (APBA) was functionalized on the surface of detonation nanodiamond (dND), and then, the specific enrichment capacity of this material was as high as 350 mg of glycoprotein per gram of dND [172]. To enhance the dispersibility and the amount of boronic acid group on the surface of dND, Yang et al. functionalized dNDs with APBA assisted by poly-L-lysine and poly(ethyleneglycol)-diglycolic acid (PEG) with good dispersibility and plentiful boronic acid functional groups [173]. Meanwhile, boronic acid has also been functionalized on MALDI plate with various supports, including Au NPs and gold-coated silicon wafer for the on-plate enrichment of glycosylated peptides [174].

Hydrazide chemistry based nanomaterials The hydrazide chemistry-based method, another important type of N-glycoproteome enrichment strategy, was first developed by Zhang et al. [175]. The carbohydrates were firstly oxidized to aldehydes and then covalented with hydrazide groups on the support. The proteolysis step was followed to remove the nonglycosylated peptides and release the N-linked glycopeptides from the support with PNGase F for the identification of glycosylated peptides and glycosylation site. Initially, resins or gels were

NANOMATERIALS FOR THE SAMPLE PREPARATION OF PROTEIN ANALYSIS

57

applied for the immobilization of hydrazine, but these materials are difficult to be dispersed and separated, and the enrichment efficiency is low. To make the enrichment method easier and more efficient, some materials including magnetic nanoparticles and Au NPs were exploited to be modified with hydrazine groups for specific pretreatment of glycosylated proteins/peptides from biological samples [176]. Among the progress, hydrazide groups were grafted on the outside of superparamagnetic silica particles, and the coupling capacity was about 36 mg of total glycosylated protein mass per gram particles [177]. To develop a simple and rapid glycosylated protein/peptide isolation method by modifying magnetic particles with hydrazine chemistry, a hydrazine-functionalized magnetic particles (HFMP) was prepared by four different routes [178]. HFCA derivatized from adipic dihydrazide and the carboxyl-silanized magnetic particles exhibited good enrichment capacity (130  5.3 mg bovine fetuin per gram particles) and best specificity for capturing of glycoprotein. A longer spacer assisted for functionalization of hydrazine on solid supports may also decrease the steric effect and hence good capture efficiency. Further, a new type of hydrazide-functionalized core-shell magnetic nanocomposite, Fe3O4@poly(methacrylic hydrazide) (Fe3O4@PMAH), was fabricated to develop a more sensitive enrichment approach for in-depth glycoproteome research, which showed more than five times enrichment capacity compared with hydrazide resin for enrichment of standard glycopeptides [171].

Lectin affinity-based pretreatment Lectins can be specifically bind to carbohydrate residues of glycoproteins or glycolipids and have been used in isolation of biomolecules [179]. Lectin binding to the target glycan structure follows several factors including multivalency, water- and metal-stabilized structures adjacent to the binding site, and the molecular geometry. Although lectins have weak and variable affinities for glycosylated proteins (mM to μM), the lectin-based pretreatment method is highly specific. To enhance the binding affinity, binding and separation protocols to special lectin have been optimized. More than 400 lectins have been discovered from a variety of species including viruses, bacteria, plants, and animals. With the high specificity and good biocompatibility, lectin has been used for capturing specific glycan structural motifs. Generally, lectins are grafted on agarose, sepharose, silica particles, or monolith as the stationary phase of chromatography column [180]. For example, concanavalin A (Con A) is the most widely used lectin and has been employed for glycosylated protein/peptide enrichment from biological samples with specific binding to oligomannosidic, hybrid, and diantennary N-glycans by forming a boronic acid-sugar-Con A bond in a sandwich structure using. Using methyl α-D-mannopyranoside, Con A was immobilized on APBA-functionalized MNPs, which was applied for the pretreatment of glycosylated proteins. From human hepatocellular carcinoma cell line, a total of 184 glycosylated sites were detected within 172 different glycosylated peptides corresponding to 101 glycosylated proteins [181]. To improve the effectiveness of glycoprotein capture, a silica-based Con A (SiO2-ODex Con A) was synthesized to enrich glycosylated proteins/peptides using oxidized dextran as the spacer, which reduced the complexity of the synthesis and provided microenvironment to maintain the spatial configuration of Con A molecules [182]. After pretreated with SiO2-ODex Con A material from human serum, the biological samples were identified with a total of 109 glycosylation sites corresponding to 50 glycoproteins in one fraction by LC-MS/MS characterization. In contrast, 86 glycosylation sites from 45 glycoproteins have been reported in human serum using the multilectin column [183].

58

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

NANOMATERIALS FOR PROTEIN IDENTIFICATION As the connection of cell physiology and genetics, proteins provide a window into complex cellular regulatory networks. Thus the high-throughput identification of proteins in complex samples is vital in biology. Prior to protein identification and characterization by MS analysis, well-defined and reproducible peptides should be obtained by the specific enzymatic reaction in a proper time. The conventional in-solution digestion is limited by time-consuming digestion procedure, autodigestion of enzyme, sample loss during concentration, and artificial modifications, severely affecting the efficiency and the result of the identification of proteins. Therefore, developing novel digestion methods was necessary to achieve highly efficient proteolysis for the identification of proteins. Besides the conventional procedure for the identification of proteins or peptides by digestion, matrix-assisted laser desorption ionization-imaging mass spectrometry (MALDI-IMS) is another technology for generating molecular maps within the tissue section by the combination of the identification of proteins and peptides with their spatial localization [184]. Therefore, MALDI-IMS is an unbiased approach to search for potential disease-related proteins and peptides (complementing immunohistochemistry). In imaging application, matrix is coated onto the whole tissue sections, and the mass spectra signals of targets molecules are acquired and analyzed into two- or three-dimensional sections. To enhance the molecule weight field analyzed by MALDI-MSI and its sensitivity, various matrices have been developed to increase the MS signals. With the development of nanoproteomics, nanomaterials have play important roles in the above demands including enzyme immobilization and MALDI-MSI matrix.

ENZYME IMMOBILIZATION To develop more efficient digestion procedures, the immobilized-enzyme reactors have recently attracted considerable interest. Immobilized-enzyme reactors have several advantages for proteolysis in protein analysis, including a low amount of autodigestion, a larger surface and interface area for digestion, a rapid proteolysis, and the good recovery of samples. The result from experiments using these immobilized-enzyme reactors has a higher sequence coverage than that obtained using conventional in-solution digestion [185]. Immobilization of enzyme requires a stationary phase/support onto which enzyme is fixed via covalent linkage, adsorption, affinity binding, or entrapment/encapsulation [186]. In recent years, various types of nanomaterials have been employed in enzyme immobilization, such as nanoparticles, nanofibers, and mesoporous materials [187]. Nanomaterials have several advantages such as large surface areas, tunable pore sizes that can be tailored to the dimensions of proteins, readily functionalized surfaces, multiple sites for interaction or attachment, and reduced mass transfer limitation. Consequently, researchers have used nanomaterials to prepare various forms with different stationary phases for a high amount of enzyme and ultimately offering enzyme-linked materials with long-term stability and enhanced proteolytic efficiency.

Magnetic nanoparticles Magnetic nanoparticles (MNPs) can be easily dispersed in solution and recovered by a simple magnet, and these materials mitigate the shortcoming of general nanoparticles used as carriers of enzyme, such as forming clumps and sonication with ultrasonic waves for temporary dispersion and separation by centrifugation or membrane. In most cases, enzyme was immobilized on the surface of MNPs via covalent bonding via cross-linker groups including epoxide groups [188, 189], aldehyde groups [190],

NANOMATERIALS FOR PROTEIN IDENTIFICATION

59

carboxylic groups [191], and terminal amine group [192]. With the assistance of the trypsinimmobilized MNPs, the protein digestion could be completed in a short time (5 min), without any complicated reduction and alkylation procedures. Meanwhile, in accordance with the requirement for the optimal catalytic activity and enzymatic stability, the mild reaction condition was considered for the immobilization of enzyme. Activated by N-hydroxysuccinimide (NHS)-1-ethyl-3(3-dimethylaminopropyl) carbodiimide (EDAC) or NHS-1-ethyl-3-[3-dimethylaminopropyl] carbodiimide hydrochloride (EDC), the carboxylic groups on the surface of the MNPs can be bonded with the amino groups on enzyme in a mild condition [193]. To comprehensively study the general functionalization strategies for MNPs, Li et al. compared the properties of the immobilized trypsin on the MNPs with different terminal groups, such as terminal amine (–NH2), epoxy (–COC), carboxylic (–COOH), aldehyde (–CHO), thiol (–SH), and maleimide (–PMPI) groups [191]. In their investigation, the highest activity per gram of MNP was obtained on –COOH and –SH surfaces, followed by –PMPI, –NH2, and –CHO surfaces. The trypsin immobilized on the –SH surface shows efficient proteolysis to the cysteine-containing proteins. To enhance the loading amount of enzyme and decrease the steric hindrance between immobilized enzyme and proteins, a new type of approach was developed for the immobilization of enzyme on magnetic nanoparticles by a hairy non-cross-linked polymer chain [194]. The non-cross-linked hairy polymer chains have numerous binding sites and place as scaffolds to support three-dimensional enzyme immobilization. Meanwhile, as no cross-linkage is present between these polymer chains, proteins could penetrate through this material, thus leading to the improvement of the immobilized-enzyme efficiency. The physical adsorption method was normally applied for the immobilization of enzyme to decrease the loss of enzyme activity. Enzyme can be immobilized on support by ionic interactions, hydrogen-bonding networks, van der Waals forces, hydrophobic interaction, etc. As enzyme absorption does not require additional reagents, it has the advantage that this method does not affect the biological activity of enzyme. For instance, the enzyme could be bound to the MNPs via Cu2+ chelation [195, 196]. The magnetic microspheres were first modified with tetraethylorthosilicate (TEOS) and then reacted with GLYMO-IDA, formed by the reaction of GLYMO and iminodiacetic acid (IDA). Subsequently, the copper ion and trypsin were introduced onto the surface. To increase the detection sensitivity of small volume sample, the prepared microspheres were packed into the microchannel by a strong magnetic field to make an on-chip enzymatic microreactor. The proteolysis of a fraction of protein extracted from rat liver could be completed within 5 min, and 23 unique peptides corresponding to seven proteins were identified [196].

Nanoporous or mesoporous materials Based on the former MNPs, immobilization of enzyme can only occur on the surfaces. In fact, a larger surface available on the solid support will provide higher abundance of the enzyme. Therefore, porous supports are favored for the conjugation of enzymes and offer attractive loading capacity. Generally, enzymes can be immobilized into the mesoporous channels by weak physical adsorptions, such as hydrogen bonding, hydrophobic attraction, or electrostatic interaction, or by covalently linkage inside the mesoporous channels on reactive groups such as aldehyde, epoxide, and thiol groups in the nanomaterials such as SBA-15, FDU-12, and MCM-41. To achieve rapid digestion, high sequence coverage, and sensitivity in protein identification, Yang and coworkers reported protein digestion placing in the nanoreactor channels of mesoporous silica SBA-15 and evaluated the performance by MS analysis. After incubation of 10 min, eight peptides that cover 58% of the myoglobin sequence with an intense

60

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

signal (signal/noise ratio >70) were identified using the mesoporous silica-based nanoreactors. In comparison, the conventional overnight in-solution digestion of proteins generated only three peptides with 27% sequence coverage [197]. Enzyme immobilized on mesoporous structures can provide stable enzyme adsorption with high efficiency. Terracciano et al. found that the well-ordered porous array of SBA-15 (6 nm) demonstrated the greatest adsorption capacity of enzyme in the host frameworks (1 min). Meanwhile, compared with the unmodified SBA-15 with the same pore diameters, the amine-functionalized SBA-15 showed higher digestion efficiency and the proteolytic efficiency and exhibited 1000 times faster than that of using conventional in-solution method. With the confinement from molecular crowding in mesoporous pores, the catalytic activity of protein and enzyme can be both stabilized and induced order-of-magnitude enhancements. Although the adsorption of trypsin on the functionalized SBA-15 via electrostatic interaction is simple, the risk of trypsin leaching out is a concern. For covalent immobilized enzyme in the channels of mesoporous silica SBA-15, Bein et al. developed the azide-functionalized SBA-15 that reacted with acetylene-modified trypsin in a copper (I) using the “click reaction.” The resulted immobilized trypsin showed retention of enzyme activity and stability without desorption under the experimental conditions [198]. For the mesoporous material-based enzyme reactor, the proper pore size is very vital to encapsulate the enzyme inside the channels of mesoporous material for enzymatic catalysis. Meanwhile, the efficiency of size-dependent separation using mesoporous materials is based on the ratio of between pore size and the size of proteins. Thus, if the pore size is suitable for to the enzyme, the enzyme would adsorbed on the internal surface, and then the proteins smaller than the pore size will be captured in the pore channel, which is subsequently digested, while proteins larger than the pore size will be excluded. This has been applied for the development of a size-selective digestion of low-molecularweight protein method. Zou et al. covalently immobilized trypsin on the thiol-functionalized SBA15 with the pore size of ca. 5.7 nm. The materials could exclude the BSA, which has an approximate dimension of 5.0  7.0  7.0 nm3, and capture low-molecular-weight proteins including cytochrome C, lysozyme, and myoglobin and allow these small molecular weight proteins to be enzymatically digested by the immobilized trypsin [199]. By this size-selective digestion, the low-molecular-weight proteins will be separated and proteolyzed in one step, thus having the potential for the rapid proteolysis and online LC-MS analysis of low-molecular-weight proteins from complex biological samples.

Carbon based nanomaterials Owing to the high surface area, high electric conductivity, mechanical strength, and chemical stability, carbon-based nanomaterials are emerging materials that are functionalized for separation science and bioconjugation. These include carbon nanotubes, nanodiamonds, and graphene oxide. Enzyme loading capacity and efficiency of the nanomaterial are usually higher when compared with the enzyme reactors prepared from larger particles. Hereby, these have attracted attention in the immobilization of enzyme. Both single-walled carbon nanotubes (SWNT) and multiwalled carbon nanotubes (MWNT) have been used for immobilization of protein/enzyme by physical adsorption and covalent bonds. Yang and coworkers reported a carbon-based nanomaterial in which trypsin was immobilized on carbon nanotubes assisted by dendrimer spacers [200]. Further, they fabricated a trypsin-immobilized polyaniline (PA)-coated Fe3O4/carbon nanotube composite, in which CNTs can increase its specific surface area and dispersibility. Additionally, polyaniline serving as the “outer clothing” was introduced into the composite to protect the nanostructures and to accommodate trypsin [201]. The obtained

NANOMATERIALS FOR PROTEIN IDENTIFICATION

61

trypsin-immobilized PA/Fe3O4/CNT has been coupled with MALDI-TOF MS for the digestion and peptide mapping of bovine serum albumin (BSA), myoglobin, and lysozyme with the corresponding amino acid sequence coverage of 46%, 81%, and 63%, respectively. As a nanomaterial member of carbon allotropes, nanodiamond has a specific chemical stability and biocompatibility. Detonation nanodiamonds (dNDs) have a large surface area and a variety of surface functional groups, which can be utilized in immobilization of enzyme. After activated by carbodiimide and treated with NHS, the carboxylated dNDs was functionalized with trypsin by covalent bonds, and subsequently used for the proteolysis of myoglobin. These materials produced superior sequence coverage as compared with that obtained with in-solution digestion or commercial Poroszyme® beads [202]. Meanwhile, the enzymatic performance immobilized on dNDs was also investigated by the proteolysis of standard proteins, including the pH tolerance, temperature, immobilized time, and nanoparticle concentrations. The optimized nanoparticles/nanomaterials with immobilized enzymes often show four advantages than the native enzymes: (i) wider temperature tolerance range, (ii) broader working pH, (iii) greater thermostability, and (iv) reusability. The high surface-to-volume ratio and chemical, electrical, and optical properties of nanoparticles or nanomaterials can increase enzyme loading and affect the diffusion of immobilized enzymes. Possessing specific structures, graphene oxide is an ideal nanomaterial with large surface area for the immobilization of enzyme. With the help of the abundant hydrophilic groups, the immobilized GO exhibits well dispersibility in aqueous solution. Jiang et al. functionalized GO sheets with aminefunctionalized Fe3O4 nanoparticles via covalent bond [203]. Trypsin was following immobilized on the prepared material via stacking and hydrogen bonding for efficient microwave-assisted proteolysis. The novel trypsin-immobilized nanocomposite was successfully employed for the digestion of bovine serum albumin and myoglobin with the 275 μg mg1 of the enzyme immobilization amount. As the enzyme-functionalized GO is well dispersed in aqueous solution, these materials can find a wide range of applications in the fabrication of microchip bioreactors for highly efficient proteolysis. The feasibility and performance of the novel GO-based microfluidic bioreactors were demonstrated by the proteolysis of standard proteins, and the digestion time was significantly reduced to 5 s. Recently, Yang immobilized trypsin onto the GO surface assisted to fabricated bioreactors for efficient microwaveassisted proteolysis [204]. The use of spacer molecules for the immobilization of trypsin on graphene oxide proved to be beneficial. For instance, dendrimer was applied as spacer for the functionalization of trypsin onto GO nanosheets on which the loading amount of trypsin was calculated to be about 649  20 mg g1, and an efficient digestion of standard proteins could be obtained within 15 min [205].

Other nanomaterials There are other nanomaterials that either are underdeveloped or show promise in bioconjugates, such as gold nanoparticles, metal-organic frameworks (MOFs), and nanoporous alumina. Due to their unique physical and chemical properties, gold nanoparticles (Au NPs) have also attracted interest for enzyme immobilization. Yang et al. reported a microreactor established by entrapping trypsin in a microchip coated with a gold nanoparticle network for the efficient online proteolysis of low-abundance proteins and complex extracts of the mouse macrophages. The PDDA/Au NP-assembled microchip was fabricated via a layer-by-layer electrostatic binding of PDDA and Au NPs. Here, charge-stabilized Au NPs provide a mild microenvironment similar to that of enzymes in native states and thus offer a large specific surface area for enzyme binding. The maximum proteolytic rate of the immobilized trypsin was

62

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

400 mM min1 (mg of enzyme)1, and low-abundance proteins down to femtomole could be proteolyzed. Using this enzymatic microreactor, proteins isolated from the mouse macrophages were efficient digested, and 497 proteins were identified by 2-D-HPLC-ESI-MS/MS analysis. Due to their extraordinary surface areas, high porosity, and high functional tenability, MOFs have also become a promising type of porous materials for the biological conjugation [206].

LASER DESORPTION IONIZATION MASS SPECTROMETRY IMAGING (LDI-IMS) AND MATRIX Matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) and electrospray ionization mass spectrometry (ESI-MS) have played important roles in the proteomics. ESI-MS can easily couple with online pre-MS separation, such as high-performance liquid chromatography or capillary electrophoresis. Meanwhile, MALDI-MS is a robust platform for the identification of proteins due to ease of sample preparation and tolerance of salts. In MALDI-MS, low-molecular-weight and energyabsorbing matrices are used over the target samples or tissue sections to minimize sample damage from laser irradiation. The suitability of an organic compound as matrix is determined by the type of the laser and its emission wave length. The most popular of matrices for MALDI-MS with a N2 laser at 337 nm include 2,5-dihydroxybenzoic acid (DHB), a-cyano-4-hydroxycinnamic acid (CHCA), and sinapinic acid (SA). Although the above matrices are essential in the energy relay process from the laser and transfer it to crystallized target molecules, these matrix molecules are ionized and generate significance background signals of small matrix ion suppressing and hindering small-molecule analysis. Although a homogeneous mixture is assumed to be spotted on the MALDI plate, “hot spots” exist on the plate with traditional matrix and thus limited the record of uniform signal of LDI-IMS. Therefore, nanomaterials of various sizes, shapes, and compositions have been tested for their applicability in the MS matrix for analysis of targets in aqueous sample or tissue section to solve the limitation of traditional matrices [207]. Nanomaterials have been applied and designed as efficient assisted matrices for LDI-MS with their unique properties including large surface area, absorption in the ultraviolet region (220–370 nm), and abundance functional group. Compared with the traditional organic matrices, nanomaterial-assisted LDI has some advantages including the accessible detection of the low-molecule-weight regions (<700 Da) with a background-free mass spectrum, the excellent shot-to-shot reproducibility, the enhancement of signal intensity, and the ability of pretreatment of analytes from complex samples. Nanomaterials including carbon-based and metal-based materials have been successfully applied as the assisted matrices in LDI analysis.

Carbon-based nanomaterials Carbon-based nanomaterials including carbon nanotube (CNTs), fullerenes, nanodiamond, and graphene have been used as promising matrices for LDI to analyze proteins, peptides, and corresponding small molecules. CNT was firstly applied as an effective MALDI matrix for small molecules in 2003 [208]. In the related study, oxidized CNT was reported to be a matrix substance for the analysis of small molecules, oligosaccharides, peptides, and insulin [209, 210]. Due to the oxidation process, many impurities and surface functionalization were removed on CNTs. The oxidized CNT matrix thus exhibits excellent reproducibility of peak intensity within and between sample spots. Additionally, intensity and signal-to-noise ratios of analytes are much improved over the simple CNTs. Furthermore, CNTs can be

NANOMATERIALS FOR PROTEIN IDENTIFICATION

63

functionalized to be both the adsorbent in solid-phase extraction and the matrix for LDI-MS for the analysis of small molecules including β-Arg, propranolol, and Leu-Tyr. The detection limit for small molecules improved 10–100-fold when the samples were pretreated by CNTs [211]. With the high thermal conductivity and transparent in near-UV region, detonation diamond has also gained attention as the matrix for the MALDI- or imaging MS. Detonation nanodiamond with the diameter from 5 to 10 nm can be deposited as a thin layer on MALDI plate to enhance the ionization of compounds based on the combined characteristics of physisorption and chemisorption on the detonation nanodiamond surfaces. Furthermore, Wang et al. prepared the sample using three partially separated layers formed by matrix, diamond nanoparticles, and analyte solutions. This method compensated the difference in ionization efficiency between carbohydrates and proteins and allowed the direct analysis of carbohydrates from protein mixtures [212]. With subbandgap absorption and heat confinement inside the wires, the boron-doped diamond nanowires (BDD NWs) were applied as the inorganic substrate for matrix-free laser desorption/ionization mass spectrometry (LDI-MS) analysis of small molecules, on which the limit of detection (LOD) of verapamil was 200 zmol μL1 [213]. With the excellent optical and electric properties, graphene has been acted as an excellent matrix for LDI-MS [214]. Compared with traditional organic matrix, graphene exhibited a high desorption/ ionization efficiency for nonpolar compounds. Meanwhile, graphene as matrix could avoid fragmentation of analytes and offer good reproducibility and salt tolerance. Min et al., prepared various types of different surface-immobilized carbon materials utilizing graphene oxide (GO), reduced GO (rGO), multiwalled CNT (MWCNT), and aminated MWCNT for comparison of LDI efficiency. GO/ MWCNT-NH2 and rGO/MWCNT-NH2 double-layer platforms showed excellent performance for the analysis of enzyme activity, small molecules, and mouse brain tissue mass imaging because of negligible interference, high salt tolerance, mechanical durability, and homogenous signal intensities across regions [215]. To enhance the ionization efficiency and sensitivity of LDI-MS, the composites of graphene combined with other materials were developed, such as graphene coated with mesoporous silica (G@SiO2) and graphene oxide embedded sol-gel (GOSG) film [216, 217].

Metal-based nanomaterials Metal-based nanomaterials are also important materials that can be used as highly efficient matrices for the biological molecule analysis by LDI-MS, such as Au NPs [218], silver nanoparticles [219], TiO2 [220], zinc oxide (ZnO) nanowires [221], platinum nanoflowers [222], ZrO2 [223], and BaTiO3 nanoparticles [224]. These nanomaterials provide several advantages that include a large surface-areato-volume ratio, strong absorption in UV spectral range, high stability, and operability. Due to the straightforward sample preparation, high ionization coefficiency, excellent stability, and biocompatibility, Au NPs have achieved the high attention as potential inorganic matrix for the analysis of small molecules by LDI-MS. Russell et al. firstly applied size-selected (2–10 nm) Au NPs for the MALDI analysis of peptides. However, with the production of Au cluster peaks, the application of Au NPs was limited in LDI-MS [225]. To solve the above problem, various modified Au NPs were fabricated and used as matrices for LDI-MS. Citrate-capped Au NPs served as matrix for the determination of biomolecules including neutral steroids and carbohydrate. To avoid the poor reproducibility of Au NPs homogeneously distributed on the plate, the on-chip patterned Au NP microarray modified with a nanoscale silicate coating layer by layer was prepared and applied for analysis of small and medium compounds with LDI-TOF MS, which could be performed over many cycles with good reproducibility spot to spot [218]. With tree- or bouquet-like morphology with distinct physicochemical properties,

64

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

flowerlike Au NPs were applied to improve LDI-MS by enhancing the ionization of peptides in the mass range up to 2500 Da and providing in parallel an effective internal calibration of mass spectra. Thus, flowerlike Au NPs, with gold monoisotopicity and excellent chemical stability, have a promising feature for the quantitative and qualitative analysis of putative biomarkers in diagnosis and therapy of diseases. TiO2 nanomaterials exhibit strong UV absorption characteristics with the 3.2 eV band gap of bulk anatase and the capability to rapidly transmit energy to analytes. Therefore, it was potentially applied as matrices for the analysis of peptides or small proteins by LDI-MS. TiO2 nanocrystals (TiO2 nanoparticles (TiO2 NPs)), TiO2 nanotubes (TiO2 NTs), and TiO2 prolate nanospheroids (TiO2 PNSs) with different shapes and sizes were investigated for the quantitative analysis of peptides or small proteins (steroid hormones, amino acids, and saccharides). It showed that all TiO2 nanocrystals, regardless of their size and shape, could be used as substrates for the efficiency ionization of steroid hormones, amino acids, and saccharides. Meanwhile, the larger nanocrystals, TiO2 PNSs and TiO2 NTs, exhibit the best reproducibility having potential applications for the quantification of small molecules [226]. Boukherroub et al. reported a method for analysis of peptides and small molecules by using surfaceassisted laser desorption-ionization mass spectrometry (SALDI-MS) coated with a titanium oxide (TiO2) nanotube layer based on electrochemical anodization. Compared with the classical MALDIMS, the detection limit was improved 10-fold for the analysis of tyrosine kinase inhibitor and verapamil with the prepared TiO2 nanotube substrate [227]. Moreover, other TiO2 nanocomposites including nylon nanoweb with TiO2 nanoparticles and surface-modified BaTiO3 nanoparticles were also prepared as matrix of LDI-MS. The novel matrix can spread on the target plate homogeneously using a nylon nanoweb as a solid scaffold. With leucine-enkephalin (555.6 g mol1) and cyclic citrullinated peptide (1668 g mol1) as model analytes, the intensities of mass peaks and the S/N ratio of mass spectra exhibited to be far more improved than that observed with the TiO2 nanoparticles without nanowebs [228]. Nowadays, owing to the large surface area, tunable pore sizes, high porosity, and UV-range absorption, metal-organic frameworks (MOFs) have been used as matrix for LDI-MS. In the series MOFs, the cage-type MIL-100(Fe) gives a good stability of MS intensity and a good response with the polycyclic aromatic hydrocarbons (PAHs) [229]. Further, they applied MIL-100(Fe) as matrices for the LID-MS analysis of polar carbohydrates (glucose; sucrose; galactose; lactose; fructose; maltose; and a-, b-, and g-cyclodextrins (a-, b-, and g-CD)) with good reproducibility and low background interferences [230]. Combined the merits of MOFs with magnetic particles, magnetic nanocomposites coated with ZIF-8 (Fe3O4@ZIF-8 MNCs) were fabricated and used as pretreatment probe and matrix for the LDI-MS analysis of small molecules [231]. Recently, Huang et al. prepared carbonized MOFs (cMIL-53 and cCYCU-3) and applied these materials for the analysis of carbohydrates, peptides, PAHs, and phenolic acids with better MS response than the pure MOFs and nanoporous carbons as matrix [232].

NANOMATERIALS FOR THE QUANTIFICATION OF PROTEIN Proteins are vital biomolecules in living organisms and are closely related to the discovery of biomarkers in diseases. Meanwhile, the quantitative expression of proteins and their interacting partners may help systematic analysis of biological processes. Hence, highly efficient determination of absolute protein amounts and the quantification of differentially expressed proteins are one of the key subjects in

NANOMATERIALS FOR THE QUANTIFICATION OF PROTEIN

65

proteomics analysis. Despite great advancements in the tools for the detection and quantification of proteins, the analysis of low-abundance proteins still faces great challenges. Nanomaterials with desired properties have been optimized for detection and quantification of low-abundance proteins. Herein, recent advances using different nanomaterials for the detection and quantification of proteins in an organism were summarized, with particular emphasis on the outlook for different nanomaterials applied for the quantitative protein analysis, as well as possibilities and limitation of nanomaterials in this rapidly growing field.

NANOMATERIALS FOR PROTEIN MICROARRAY In the systematic analysis of biological processes, the quantitative expression of proteins and their interacting proteins is a vital component in proteomics research. The differential expression of proteins, suggested by quantitative proteomics, reveals fruitful information regarding the changes between the normal state and the disease state. As a promising technique, protein microarrays are able to directly measure changes in the proteome. Moreover, the protein microarray can composite, quantify, and characterize proteins or proteoforms in the proteome arising from posttranslational modifications, splicing events, and interactions [232]. However, the sensitive and reliable detection of protein microarray needs to be improved to meet the increasing demands in studying the large number of proteins. To quantify target proteins and the disease biomarkers in biological samples, various biosensors have been developed [233]. Additionally, label-based systems like fluorescence, chemiluminescence, and radioisotope have witnessed substantial advancements. More recently, nanomaterials such as gold nanoparticles, quantum dots, dye-doped nanoparticles, and silica nanoparticles are employed for protein quantification studies. To reduce the impediment of labeling reaction in label-based systems, label-free detection techniques, including surface plasmon resonance (SPR), nanocomposite and microcantilevers have also advanced in measuring the inherent properties of target molecules, such as mass, dielectric, or optical properties. With unique properties, nanomaterials have extended surface for immobilization of biomolecules resulting in increased binding properties and enhanced detection sensitivity. Therefore, nanomaterials are developed and incorporated in several novel detection platforms discussed in this section.

Gold nanoparticles (GNPs) With a number of unique physical and chemical properties, gold nanoparticles (Au NPs) have drawn attention as ideal candidates for sensitive detection of biomolecules. These nanoparticles are known to have excellent, electronic properties, optoelectronic property, chemical stability, high quantum efficiency in a wide range of wavelengths, and excellent biocompatibility. Moreover, these properties of Au NPs can be easily adjusted by their size, shape, interparticle distance, and the solvent and temperature of the surrounding environment. Therefore, Au NPs have been employed for the development of novel detection methods with high sensitivity, stability, and reliability [233]. As feasible alternatives to fluorophore-based probes, Au NP conjugates have been used to scatter light and enhance the sensitivity of the assay with the amplification by electroless metal deposition. Recognition antibodies functionalized on gold NPs were applied as a tool for multiplexed detection of tumor markers for lung cancer with increased sensitivity on protein microarray. Using this method, detection of biomarkers in serum samples could reach to a concentration of <1 ng mL1 in 1 h [234]. Based on an adsorption-controlled kinetic model, semi-quantitative sandwich nanoparticle

66

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

immunoassays were developed utilizing a platform that includes antibody conjugated with gold nanoparticles, whose signal is further enhanced by silver nucleation. Target analyte could be detected at a concentration as low as 0.1 μg cm3 (4 ng total) [235]. Utilizing the reduction of HAuCl4 by NH2OH and self-catalysis of Au NPs, a colorimetric method for the ultrasensitive detection of prostate-specific antigen (PSA) using a novel biotin-polyethylene glycol Au NPs probes is reported [236]. The signal of Au NPs can be amplified by introducing HAuCl4 –NH2OH solutions, in which Au NPs catalyze the reduction of Au3+ by NH2OH to form larger Au NPs. PSA and nucleic acid targets (microRNAs) from a single sample can be detected with low detection concentration (50 fM for nucleic acid targets and 1 μg L1 for the PSA target). This method shows a potential in the microarray for biological samples with high sensitivity, selectivity, and simplicity. In lectin microarray, microbe-gold nanoparticle conjugates are applied as labels to monitor specific affinity of microbes with lectins, followed by a resonant light scattering (RLS) enhancement step based on the electroless deposition of silver onto the gold particles [237]. Using the same detection principle, a microarray format for the detection of carbohydratelectin or glycoprotein-lectin interactions by gold nanoparticle probes has also been developed by Wang et al. Compared with the conventional fluorescence-based microarray format, the RLS-based microarray format exhibits lower detection limit for both carbohydrate and lectin. However, this assay is only applicable to specific carbohydrate motifs rather than the entire glycome. Furthermore, Au NPs have also been used as signal enhancers for Raman scattering-based detection of biomolecules [238, 239]. For example, in a sandwich-type SERS immunoassay, the primary antibody was attached to Au NPs, which were immobilized on a substrate. After being exposed to a solution containing PSA, the immunoassay was interacted with the secondary antibody conjugated to Au NPs with Rhodamine 6G dye. The sensitivity of the novel immunoassays has been drastically enhanced in the SERS spectra with the detection limit of this immunoassay as low as  1 ng L1 for PSA in human serum [240]. Wang et al. established a sensitive peptides microarray-based SERS assay using biotinylated anti-phosphoserine antibodies, which were tagged with avidin-conjugated Au NPs, followed by silver deposition for Raman signal enhancement to analyze the kinase function and inhibition [241]. The result demonstrates that the H89 inhibitor has highly selective inhibition to protein kinase A.

Quantum dots With a wide spectral band gap and stable light properties, quantum dots (QDs) are fabricated by a semiconductor fluorescent core coated with another semiconductor shell. The common QDs are composed of CdSe core covered by a ZnS shell. Upon UV excitation, this kind of QDs emits light ( 20–25 nm fwhm), which is adjusted with the size of the CdSe core. Meanwhile, the colloidal chemistry is well studied that allows the synthesis of five to six spectrally distinct colors of emission across the visible spectrum, making them excellent labels for multiplexed, high-throughput screening. QDs exhibit bright fluorescence (20–100 times higher) [242], excellent photostability against photobleaching (50–1000 times) [243], multifluorescence excitation, and greater quantum yield. Hence, with these advantages, QDs have diverse applications in diagnostic imaging [244], detection of cancer biomarkers [245], direct probing of human serum [246], and tumor biopsy analysis [246, 247]. With above advantages, QDs are applied to label peptides, protein, or oligonucleotides for the sensitive detection of microarrays [248]. In the potential Alzheimer’s disease biomarker screening research, the performance of QDs and the fluorescent dye Alexa 647 were compared. The QDs were shown as more effective reporters in the microarray, in which the limit of detection of apolipoprotein E by QDs was five times more sensitive than that of Alexa microarray [249]. Zhukov et al. innovatively

NANOMATERIALS FOR THE QUANTIFICATION OF PROTEIN

67

established the streptavidin QD-biotinylated detector antibody model, in which six cancer-specific cytokines were successfully detected down to picomolar [246]. Moreover, after pegylation, streptavidinconjugated and pegylated QDs have improved sensitivity and specificity compared with their nonpegylated form in assays analyzing cellular protein extracts [250]. Combining the planar and suspension microarray, Jia et al. fabricated a sensitive and selective microbead array for multiplexed detection of three lung cancer biomarkers including carcinoembryonic antigen (CEA), fragments of cytokeratin 19 (CYFRA21-1), and neuron-specific enolase (NSE). QDs functioned as fluorescent signal amplifiers that result in low detection limit (CEA, 0.19 ng/mL; CYFRA21-1, 0.97 ng/mL; NSE, 0.37 ng/mL; and S/N ¼ 3). These probes can also be used directly with serum [251]. Recently, Sukhanova et al. designed a novel multiplexed suspension immunoassay system based on QD-encoded microspheres for simultaneous detection of three lung cancer markers in the human bronchoalveolar lavage fluid (BALF). The system showed almost identical results utilizing Luminex xMAP® assay [252]. However, the QDs conjugation and detection steps make it difficult to establish the best labeling technology and widespread application. To extend the QDs applied in the biology, several novel QD styles have been fabricated and applied for the rapid and high-throughput biological application, such as CdSeTe/CdS QDs [253], QD-encoded hydrogel microparticles [254], and Mn-doped ZnS QDs [255].

Other nanomaterials Due to the distinct electric and spectroscopic properties with strong and characteristic resonance Raman signatures, single-walled carbon nanotubes (SWNTs) have been applied as a promising label for SERS-based protein detection [256]. SWNTs possess unique properties such as enormous Raman scattering cross section (1021 cm2 sr1 molecule1) and simple and tunable spectra. Dai et al. functionalized macromolecular SWNTs with specific Raman-labeled antibodies as multicolor microarray slide. The SWNT tags enable strong Raman intensity and decrease the detection limitation of protein down to 1 fM, which is three orders of magnitude more sensitive than conventional fluorescence [257]. Recently, Strano et al. developed a sensitive and selective label-free protein detection platform on the coupling of aptamer-anchor polymers to semiconducting SWNT near-infrared (nIR) emitters [258]. The selectivity for specific protein was realized via synthetic DNA aptamers adhered to SWNTs. On the platform, the single secreted proteins are from organisms including Escherichia coli, HEK 293, and Pichia pastoris cells in real time. Combined with the selectivity provided by aptamers, SWNTs are a promising nanomaterial for the long-term optical monitoring of specific protein from single cells over long timescales. Meanwhile, other carbon-based nanomaterials are also prepared as biosensors for the detection of biomarkers [259]. Dye-doped silica nanoparticles (NPs) are a type of silica nanoparticles that trap a large quantity of fluorescent dye molecules inside the silica matrix. This design leads to an increase of the quantum yield of the fluorophores, thus enhancing its brightness [260]. Silica NPs are known for their minimal toxicity, biocompatibility, environment-friendliness, easy modification and high recovery rate, and resistance to microbial growth. Therefore, entrapment of dye molecules in silica NPs enables selectively labeling on some important sample such as cancer cells, bacteria, and specific targets [261]. Rhodamine B isothiocyanate-doped silica NPs were synthesized using the St€ober method, and these molecules have an average fluorescence intensity that is 4000 times higher than conventional organic dye molecules. The novel dye-doped silica NPs were also used for the sensitive detection of reverse-phase protein microarrays [262]. Fluorescein-doped silica NPs modified with glycans were

68

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

used as labels for screening carbohydrate targets on lectin super microarray. Compared with other traditional techniques, the rapid and high-throughput nature of the described lectin super microarrays, in conjunction with the versatility of the nanoparticle-based glycan labeling technique, provides a robust and unique approach for quantitative glycan profiling in a large library [263]. These novel platforms open new opportunities for fundamental research in glycobiology and for the development of novel diagnostic tools.

NANOMATERIALS FOR INDUCTIVELY COUPLED PLASMA MASS SPECTROMETRY (ICP-MS) As an element-specific detector, inductively coupled plasma-mass spectrometry (ICP-MS) has the advantages including low detection limits, good tolerance to sample matrix, a wide range of linear response, high sensitivity, and the ability of multiplex detection. Coupled with the specificity of the antigen-antibody assay, ICP-MS-based immunoassay has become a revolutionary technique for biomolecule quantification including protein [264–266], DNA [267, 268] and RNA [269, 270], living cells [271, 272], and bacteria [273, 274] with perfect sensitivity and excellent accuracy. Naturally, some special heteroatoms including metal isotopic (e.g., iron, zinc, and copper) and nonmetal atoms (e.g., phosphorus, sulfur, and selenium) present in biomolecules, which can be directly quantification by ICP-MS. However, the absence of metal atoms, in addition to existing challenges such as high detection limits and signal interference of the endogenous phosphorous and sulfur tags, limits the ICP-MS development. To overcome it, several types of element tags, including metal chelates, metal-containing compounds, and polymer-based elemental tags, have been applied for the quantification targets by ICPMS. For example, the metal-containing NPs were applied as element tags to increase their sensitivity in ICP-MS. Here, we summarized the application of metal nanomaterials in the quantification of proteins by ICP-MS. With the specificity of the antibody, the ICP-MS-based immunoassay does not require chromatographic separation and then easily reduces the effect of complex biological matrix. Zhang et al. firstly developed the ICP-MS immunoassay with DTTA chelate labeling workflow for the quantification of the thyroid-stimulating hormone (TSH) in serum [275]. To improve specific identification and signal amplification of ICP-MS immunoassay, different types of metal nanoparticles were utilized including Ag NPs, Au NPs, PbS NPs, quantum dots, metal-doped silica NPs, and upconversion NPs (Table 2.4). With its unique properties, Au NPs are the most used nanomaterials in the quantification of proteins by the ICP-MS. Baranov et al. applied Au cluster (<2 nm in diameter)-labeled antibodies combined with ICP-MS to quantify the target proteins. This method can detect target proteins at a concentration of 0.1–0.5 ng mL1 with a linear response to protein concentration over three orders of magnitude [277]. Yan et al. applied aptamer-modified Au NPs and antibody-modified silver NPs as specific element tags for quantification of cytochrome C and insulin, respectively [282]. Combining the magnetic microparticle separation and ICP-MS detection, the ultrasensitive method for simultaneous determination of different target proteins was established (Fig. 2.4). The method provides a promising simultaneous determination of two or more low-abundance proteins from complex sample via multielement tags. Moreover, Lv et al. established an immunoassay with the signal amplification of catalytic silver deposition on immunogold tags to detect low-abundance proteins such as human carcinoembryonic antigen in serum with low limit of detection of 0.03 ng mL1 [283]. Sanz-Medel et al. developed a novel Mn-doped ZnS QD-based immunoassay platform for highly sensitive detection of cancer biomarkers.

69

NANOMATERIALS FOR THE QUANTIFICATION OF PROTEIN

Table 2.4 Analytic Performance of ICP-MS-Based Methods With Elemental Tags for the Quantification of Protein Analytic Method ICP-MS with Au NP labeling ICP-MS with Au cluster labeling

Au NP amplification-ICP-MS

Single-particle mode ICP-MS with Au NP labeling ICP-MS with nanogold labels

ICP-MS with aptamermodified Au NPs Au NPs (immunogold-silver amplification)-ICP-MS ICP-MS with Au NP labeling ICP-MS with Au NP labeling, Cs/FITC-doped MMNPS ICP-MS with Au NP labeling ICP-MS with CdSe QD labeling

ICP-MS with CdTe QDs labeling

ICP-MS with CdSe/ZnS QDs labeling ICP-MS with Mn-doped ZnS QDs labeling

Analyte Rabbit antiIgG IgG

Linear Ranges 0.8–50 ng mL

1

LOD 0.4 ng mL

References 1

[276] [277]

0.05–1 nM

3 ng mL1 (filter immunoassay) 0.075 ng mL1 (protein sepharose A immunoassay) 0.22 ng mL1 0.0023 ng mL1 10 pM

0.0018–18 nM

0.5 pM

[279]

0.3–10 ng mL1

0.1 ng mL1

[280]

62.5–2000 pg mL1

28.7 pg mL1

[281]

0.1–20 nM 0.2–40 nM 0.07–1000 ng mL1

50 pM 110 pM 0.03 ng mL1

[282] [282] [283]

HIV-p24 CA19-9

7.5–75 pg mL1 1.5–90 units mL1

1.49 pg mL1 0.02 units mL1

[284] [285]

Caspase-3 AFP Human IgG

1–200 ng mL1 0.005–2.0 ng mL1 0.2–10 ng mL1 (Cd signal) 0.5–10 ng mL1 (Cd signal)

0.42 ng mL1 1.85 pg mL1 0.058 ng mL1 (Cd signal) 0.097 ng mL1 (Cd signal) 8 ng mL1

[286] [287] [288]

AFP HCG Human a-thrombin Human serum albumin Rabbitantihuman IgG Vascular endothelial growth factor Cytochrome c Insulin CEA

Human serum albumin Transferrin Human IgG Transferrin PSA

103–104 pg mL1

[278]

[289]

20 ng mL1 30 ng mL1 0.18 ng mL1

[290]

0.026 fg mL1

[255]

Continued

70

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

Table 2.4 Analytic Performance of ICP-MS-Based Methods With Elemental Tags for the Quantification of Protein—cont’d Analytic Method

Analyte

Linear Ranges

LOD

References

ICP-MS with CeO2-deposited mesoporous silica nanoparticles and MNPs ICP-MS with Y, Cd, Au-doped silica NPs and Cs-doped MMNPs

CEA

103–5 ng mL1

0.36 pg mL1

[291]

AFP Neuronspecific enolase (NSE) c-reactive protein (CRP) PSA

2–200 ng mL1 2–200 ng mL1

0.37 ng mL1 0.36 ng mL1

[292]

200–104 ng mL1

77 ng mL1

ICP-MS with TiO2 NPs labeling ICP-MS with PbS NPs labeling ICP-MS with NaYF4:Yb,Er upconversion nanoparticle (UCNP) labeling

1.16 fg mL1

[293]

CEA

0.2–50 ng mL1

0.058 ng mL1

[294]

AFP

0.5–35 ng mL1

0.22 ng mL1

[295]

The amplification of signal ICP-MS was achieved by the deposition of gold ions on the Mn-doped ZnS QD tags, on which gold ions might be reduced to gold. The sensitivity of detection of prostate-specific antigen (PSA) is at the low attog mL1 level. Another highly sensitive immunoassay based on singleparticle mode detected by ICP-MS in a time-resolved analysis (TRA) mode with Au NPs serving as model tags was developed. Based on the single-particle mode, the limit of detection of the ICP-MS immunoanalysis of rabbit-antihuman IgG was nearly one order of magnitude lower than traditional immunoassays based on integral mode [280]. With the development of different metal nanomaterials, the ICP-MS immunoassay has expended the quantification of low-abundance targets in complex sample. However, signal amplification, the elimination of matrix, and high-throughput detection should be taken for progress. Therefore, the new labeling tags for the absolute quantification of protein in complex sample by ICP-MS could be focused on high sensitivity, selectivity, and efficiency of the labeling process and multiplex tags for the proteome analysis and the determination to specific sites or modification.

SUMMARY AND PROSPECT The development of nanomaterials is attributed to the promising progress in the sample preparation, identification, and quantification of protein. Various kinds of nanomaterials possess unique properties including simple separation, high sensitivity, larger capacity, electrophonic, thermal, and mechanical properties. Meanwhile, these materials modified by various kinds of functional groups extend the different applications including pretreatment of low-abundance proteins or peptides and posttranslation

SUMMARY AND PROSPECT

71

Proteins

Surface-activated MMPs Block

1% BSA

Antibody−AgNPs Aptamer−AuNPs

Heat Dissociation

Magnetic separation ICPMS

FIG. 2.4 Schematic diagram showing the principle of the developed immunoassay and aptamer-based bioassay for determining two proteins by combining Au NPs and Ag NPs label, magnetic microparticle separation, and ICP-MS detection. Target proteins were immobilized on the surface of activated MMPs by glutaric dialdehyde coupling, followed by adding BSA to block the free activated sites. Aptamer-modified Au NPs and antibody-modified Ag NPs were added as probes to react with the corresponding protein. The protein-probe complex was separated from the sample mixture under a magnetic field. After washing, the protein-probe complex was heat dissociated (at 90°C). The MMPs were removed, and the Au NPs and Ag NPs in the solution were analyzed by ICP-MS (Copyright 2011, Royal Society of Chemistry).

modification proteins or peptides, immobilization of enzyme, supporting matrix for MS or IMS analysis, and the relative or absolute quantification of proteins. Nowadays, the pretreatment techniques and approaches developed for protein analysis have achieved enormous progress. Facing with complexity of biological samples, proteins possessing vital biological and medical functions are those relatively low-abundance proteins and having posttranslational modifications. For efficient enrichment of low-abundance proteins, silica-based, carbon-based, polymer-based, and metal-based nanomaterials have developed. However, the high sensitivity and 100% recovery could not realize in this field. Thus, the high adsorption and recovery method based on nanomaterials are needed for the pretreatment of low-abundance proteins in complex sample in the future. Meanwhile, to enhance the efficiency profile of low-abundance protein with low concentration in huge volume, the online enrichment methods combined with MS should be developed.

72

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

At present, facing with the single-cell analysis, several groups have set up methods to analyze the proteome of hundreds of cell sample [296]. Nanomaterials assisted for the cell pretreatment will make a promising development in the field. For the specific pretreatment of PTM proteins or peptides, the design and synthesis of new nanomaterials with functional groups have gained increasing attention in recent years. With various kinds of PTM in biology, most of nanomaterials focused on the two major forms of protein PTM, namely, phosphorylation and glycosylation. Because of the hydrophilic group of glycosylation and phosphorylation, two of them can be enriched by some pretreatment nanomaterials on time, which would give chance to the simultaneous characterization of cross talk of them. However, in the specific pretreatment method, both selectivity and sensitivity are indispensable. The ideal pretreatment method for PTM enrichment is to reach a balance between selectivity and sensitivity, which present high demand to the fabrication and design of nanomaterials. Meanwhile, the challenges of revealing other PTMs still demand the development of new approach and novel nanomaterials for the enrichment and identification of them. For the efficient identification of proteins in biological sample, nanomaterials combined with MS have made a stride in the deep profiling of the proteome. The immobilization of enzyme on different nanomaterials has shown improved proteolysis resulting higher sequence coverage and long-term enzyme stability, in addition to possible reusability of the nanomaterials. Recently, coupling the enzymeimmobilized nanomaterials with MS and/or LC is applicable to the development of automated, high-throughput proteomics analyses. With the demand of deep profile of proteome and PTM analysis, novel pretreatment methods of online sample extraction, digestion, enrichment, and separation shed light on designing and synthesizing new materials. Additionally, these novel nanomaterials with unique properties, namely, large surface area, strong ultraviolet absorption, and special optical properties, have enabled these materials to be ideal matrices for the analysis of different kinds of targets (including small molecules, peptides, and proteins) and imaging mass spectrometry analysis. These nanomaterialbased matrices have empowered a more robust mass spectrometry-based analysis. The biological samples pretreated by nanomaterials are more homogenous than traditional methods and thus eliminate the presence of “sweet spots". The experimental workflow also reduces background signals in the low mass region. However, the inorganic matrices capable of analyzing high-molecular-weight proteins are limited. Additionally, internal standard in the imaging mass spectrometry is need for the quantification of biological molecules for MS imaging. In the systematic analysis of biological processes, relative and absolute quantification provides vital information regarding the relationship among proteins, protein families, and complexes. In recent years, protein microarray offers many avenues to simultaneously analyze multiple samples to achieve relative quantitation of proteins for potential use in diagnosis, prognosis of diseases, and possible therapeutic efficacy. To improve the sensitivity and quality of microarray data, different nanoparticle amplifiers such as Au NPs, QDs, CNTs, and dye-doped silica NPs have been developed to bind target of interest. In the future, standardization efforts concerning experimental design of the nanoparticle amplifier and the assay itself and quality controls are stringently required to compare the results produced in different laboratories. Meanwhile, the novel nanoparticles that possess good stability in aqueous and high ionic solution will benefit for the reliable and reproducibility of protein microarray data. As nanomaterials have great potential in the field of label-free assays, new nanomaterials are being developed by studying the surface chemistry of nanoparticle. Finally, the development of multifunctional nanomaterials will extend their future utility as signal amplifiers in protein microarrays. ICP-MS has drawn more attention with the development of labeling strategies and nanomaterial tags for the quantification

REFERENCES

73

proteins, nucleic acid, cells, bacteria, and viruses. In the future, the ICP-MS assisted by nanomaterials will continue to evolve in the design and fabrication of novel tags. Meanwhile, to keep pace with proteome, the accuracy of the quantification for multiplex detection in complex samples should be ensured in the ICP-MS with labeling tags. Furthermore, for the application of ICP-MS in the quantification of a smaller size organism, novel element tags are needed to enhance the synergy of ICP-MS and other technologies including luminescence imaging and molecular MS. With the development of novel element tags, ICP-MS will continue to play an important technique in the further clinical research.

ACKNOWLEDGMENT This work is supported by Special Project on Precision Medicine under the National Key R&D Program (SQ2017YFSF090210), National Key Research and Development Program of China (2016YFA0501300, 2017YFA0505100, and 2017YFA0505001), National NSF (21305018, 31570825, and 31600665), RFDP (20130071140007), Shanghai Pujiang Program (18PJD002) and Open Foundation of Shanghai Key Laboratory of Forensic Medicine (KF1404). We would like to thank Zhijie Wu for critical reading of this manuscript.

REFERENCES [1] Cafarelli TM, Desbuleux A, Wang Y, Choi SG, De Ridder D, Vidal M. Mapping, modeling, and characterization of protein–protein interactions on a proteomic scale. Curr Opin Struct Biol 2017;44:201–10. [2] Chiharu U, Masatoshi K. RING-, HECT-, and RBR-type E3 ubiquitin ligases: involvement in human cancer. Curr Cancer Drug Targets 2016;16:157–74. [3] Coates CJ, Decker H. Immunological properties of oxygen-transport proteins: hemoglobin, hemocyanin and hemerythrin. Cell Mol Life Sci 2017;74:293–317. [4] Hasanpourghadi M, Pandurangan AK, Mustafa MR. Modulation of oncogenic transcription factors by bioactive natural products in breast cancer. Pharmacol Res 2017;128:376–88. [5] Subbaiah VK, Kranjec C, Thomas M, Banks L. PDZ domains: the building blocks regulating tumorigenesis. Biochem J 2011;439:195–205. [6] Huang G, Willems K, Soskine M, Wloka C, Maglia G. Electro-osmotic capture and ionic discrimination of peptide and protein biomarkers with FraC nanopores. Nat Commun 2017;8:935. [7] Kuenzi BM, Remsing Rix LL, Stewart PA, Fang B, Kinose F, Bryant AT, et al. Polypharmacology-based ceritinib repurposing using integrated functional proteomics. Nat Chem Biol 2017;13(12):1222. [8] Casadevall D, Kilian AY, Bellmunt J. The prognostic role of epigenetic dysregulation in bladder cancer: a systematic review. Cancer Treat Rev 2017;61:82–93. [9] Wilkins M. Proteomics data mining. Expert Rev Proteomics 2009;6:599–603. [10] Noble D. Modeling the heart—from genes to cells to the whole organ. Science 2002;295:1678–82. [11] Aranda S, Mas G, Di Croce L. Regulation of gene transcription by Polycomb proteins. Sci Adv 2015;1: e1500737. [12] Postic G, Ghouzam Y, Chebrek R, Gelly J-C. An ambiguity principle for assigning protein structural domains. Sci Adv 2017;3:e1600552. [13] Goyama S, Huang G, Kurokawa M, Mulloy JC. Posttranslational modifications of RUNX1 as potential anticancer targets. Oncogene 2014;34:3483. [14] Marcotte EM, Pellegrini M, Ng H-L, Rice DW, Yeates TO, Eisenberg D. Detecting protein function and protein-protein interactions from genome sequences. Science 1999;285:751–3. [15] Kudo A. Introductory review: periostin—gene and protein structure. Cell Mol Life Sci 2017;74:4259–68.

74

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

[16] Larsen SC, Sylvestersen KB, Mund A, Lyon D, Mullari M, Madsen MV, et al. Proteome-wide analysis of arginine monomethylation reveals widespread occurrence in human cells. Sci Signal 2016;9: rs9-rs. [17] Smith LM, Kelleher NL, The Consortium for Top Down P. Proteoform: a single term describing protein complexity. Nat Methods 2013;10:186. [18] Pagel O, Loroch S, Sickmann A, Zahedi RP. Current strategies and findings in clinically relevant posttranslational modification-specific proteomics. Expert Rev Proteomics 2015;12:235–53. [19] Zimmerman LJ, Li M, Yarbrough WG, Slebos RJC, Liebler DC. Global stability of plasma proteomes for mass spectrometry-based analyses. Mol Cell Proteomics 2012;11:1–12. [20] Tan C, Cao X, Wu X-J, He Q, Yang J, Zhang X, et al. Recent advances in ultrathin two-dimensional nanomaterials. Chem Rev 2017;117:6225–331. [21] Kong F-Y, Zhang J-W, Li R-F, Wang Z-X, Wang W-J, Wang W. Unique roles of gold nanoparticles in drug delivery, targeting and imaging applications. Molecules 2017;22:1445. [22] Zhang S, Geryak R, Geldmeier J, Kim S, Tsukruk VV. Synthesis, assembly, and applications of hybrid nanostructures for biosensing. Chem Rev 2017;117:12942–3038. [23] Xie N, Liu S, Yang X, He X, Huang J, Wang K. DNA tetrahedron nanostructures for biological applications: biosensors and drug delivery. Analyst 2017;142:3322–32. [24] da Costa JP, Oliveira-Silva R, Daniel-da-Silva AL, Vitorino R. Bionanoconjugation for proteomics applications—an overview. Biotechnol Adv 2014;32:952–70. [25] Mandal A, Singha M, Addy PS, Basak A. Laser desorption ionization mass spectrometry: recent progress in matrix-free and label-assisted techniques. Mass Spectrom Rev 2017. [26] Higashi T, Ogawa S. Isotope-coded ESI-enhancing derivatization reagents for differential analysis, quantification and profiling of metabolites in biological samples by LC/MS: a review. J Pharm Biomed Anal 2016;130:181–93. [27] Zhang YH, Wang XY, Shan W, Wu BY, Fan HZ, Yu XJ, et al. Enrichment of low-abundance peptides and proteins on zeolite nanocrystals for direct MALDI-TOF MS analysis. Angew Chem Int Ed 2005;44:615–7. [28] Zuo C, Yu W, Zhou X, Zhao D, Yang P. Highly efficient enrichment and subsequent digestion of proteins in the mesoporous molecular sieve silicate SBA-15 for matrix-assisted laser desorption/ionization mass spectrometry with time-of-flight/time-of-flight analyzer peptide mapping. Rapid Commun Mass Spectrom 2006;20:3139–44. [29] Chen W-Y, Wang L-S, Chiu H-T, Chen Y-C, Lee C-Y. Carbon nanotubes as affinity probes for peptides and proteins in MALDI MS analysis. J Am Soc Mass Spectrom 2004;15:1629–35. [30] Du Z, Yu Y-L, Yan X-R, Wang J-H. Isolation and pre-concentration of basic proteins in aqueous mixture viasolid-phase extraction with multi-walled carbon nanotubes assembled on a silica surface. Analyst 2008;133:1373–9. [31] Tian R, Zhang H, Ye M, Jiang X, Hu L, Li X, et al. Selective extraction of peptides from human plasma by highly ordered mesoporous silica particles for peptidome analysis. Angew Chem Int Ed 2007;46:962–5. [32] Tian R, Ren L, Ma H, Li X, Hu L, Ye M, et al. Selective enrichment of endogenous peptides by chemically modified porous nanoparticles for peptidome analysis. J Chromatogr A 2009;1216:1270–8. [33] Deng Y, Qi D, Deng C, Zhang X, Zhao D. Superparamagnetic high-magnetization microspheres with an Fe3O4@SiO2 core and perpendicularly aligned mesoporous SiO2 shell for removal of microcystins. J Am Chem Soc 2008;130:28–9. [34] Chen H, Liu S, Yang H, Mao Y, Deng C, Zhang X, et al. Selective separation and enrichment of peptides for MS analysis using the microspheres composed of Fe3O4@nSiO2 core and perpendicularly aligned mesoporous SiO2 shell. Proteomics 2010;10:930–9. [35] S-f R, Guo Y-l. Carbon nanotubes (2,5-dihydroxybenzoyl hydrazine) derivative as pH adjustable enriching reagent and matrix for MALDI analysis of trace peptides. J Am Soc Mass Spectrom 2006;17:1023–7. [36] Chen X, Wang W, Song Z, Wang J. Chitosan/carbon nanotube composites for the isolation of hemoglobin in the presence of abundant proteins. Anal Methods 2011;3:1769–73.

REFERENCES

75

[37] Zhang Z, Yang X, Chen X, Zhang M, Luo L, Peng M, et al. Novel magnetic bovine serum albumin imprinted polymers with a matrix of carbon nanotubes, and their application to protein separation. Anal Bioanal Chem 2011;401:2855. [38] Shiea JT, Huang JP, Teng CF, Jeng JY, Wang LY, Chiang LY. Use of a water-soluble fullerene derivative as precipitating reagent and matrix-assisted laser desorption/ionization matrix to selectively detect charged species in aqueous solutions. Anal Chem 2003;75:3587–95. [39] Vallant RM, Szabo Z, Bachmann S, Bakry R, Najam-ul-Haq M, Rainer M, et al. Development and application of C60-fullerene bound silica for solid-phase extraction of biomolecules. Anal Chem 2007;79:8144–53. [40] Vallant RM, Szabo Z, Trojer L, Najam-ul-Haq M, Rainer M, Huck CW, et al. A new analytical materialenhanced laser desorption ionization (MELDI) based approach for the determination of low-mass serum constituents using fullerene derivatives for selective enrichment. J Proteome Res 2007;6:44–53. [41] Chen H, Qi D, Deng C, Yang P, Zhang X. Preparation of C60-functionalized magnetic silica microspheres for the enrichment of low-concentration peptides and proteins for MALDI-TOF MS analysis. Proteomics 2009;9:380–7. [42] Chen H, Xu X, Yao N, Deng C, Yang P, Zhang X. Facile synthesis of C8-functionalized magnetic silica microspheres for enrichment of low-concentration peptides for direct MALDI-TOF MS analysis. Proteomics 2008;8:2778–84. [43] Yao N, Chen H, Lin H, Deng C, Zhang X. Enrichment of peptides in serum by C8-functionalized magnetic nanoparticles for direct matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis. J Chromatogr A 2008;1185:93–101. [44] Chen H, Deng C, Li Y, Dai Y, Yang P, Zhang X. A facile synthesis approach to C8-functionalized magnetic carbonaceous polysaccharide microspheres for the highly efficient and rapid enrichment of peptides and direct MALDI-TOF-MS analysis. Adv Mater 2009;21:2200–5. [45] Huang LCL, Chang HC. Adsorption and immobilization of cytochrome c on nanodiamonds. Langmuir 2004;20:5879–84. [46] Chen W-H, Lee S-C, Sabu S, Fang H-C, Chung S-C, Han C-C, et al. Solid-phase extraction and elution on diamond (SPEED): a fast and general platform for proteome analysis with mass spectrometry. Anal Chem 2006;78:4228–34. [47] Kong XL, Huang LCL, Hsu CM, Chen WH, Han CC, Chang HC. High-affinity capture of proteins by diamond nanoparticles for mass spectrometric analysis. Anal Chem 2005;77:259–65. [48] Wei L-M, Shen Q, Lu H-J, Yang P-Y. Pretreatment of low-abundance peptides on detonation nanodiamond for direct analysis by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. J Chromatogr B 2009;877:3631–7. [49] Geim AK. Graphene: status and prospects. Science 2009;324:1530–4. [50] Liu J-W, Zhang Q, Chen X-W, Wang J-H. Surface assembly of graphene oxide nanosheets on SiO2 particles for the selective isolation of hemoglobin. Chem Eur J 2011;17:4864–70. [51] Yin P, Wang Y, Li Y, Deng C, Zhang X, Yang P. Preparation of sandwich-structured graphene/mesoporous silica composites with C8-modified pore wall for highly efficient selective enrichment of endogenous peptides for mass spectrometry analysis. Proteomics 2012;12:2784–91. [52] Yin P, Zhao M, Deng C. High efficiency enrichment of low-abundance peptides by novel dual-platform graphene@SiO2@PMMA. Nanoscale 2012;4:6948–50. [53] Cheng G, Liu Y-L, Wang Z-G, Zhang J-L, Sun D-H, Ni J-Z. The GO/rGO-Fe3O4 composites with good water-dispersibility and fast magnetic response for effective immobilization and enrichment of biomolecules. J Mater Chem 2012;22:21998–2004. [54] Liu Q, Shi J, Cheng M, Li G, Cao D, Jiang G. Preparation of graphene-encapsulated magnetic microspheres for protein/peptide enrichment and MALDI-TOF MS analysis. Chem Commun 2012;48:1874–6. [55] Zhang P, Fang X, Yan G, Gao M, Zhang X. Highly efficient enrichment of low-abundance intact proteins by core-shell structured Fe3O4-chitosan@graphene composites. Talanta 2017;174:845–52.

76

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

[56] Jia WT, Chen XH, Lu HJ, Yang PY. CaCO3-poly(methyl methacrylate) nanoparticles for fast enrichment of low-abundance peptides followed by CaCO3-core removal for MALDI-TOF MS analysis. Angew Chem Int Ed 2006;45:3345–9. [57] Xiong H-M, Guan X-Y, Jin L-H, Shen W-W, Lu H-J, Xia Y-Y. Surfactant-free synthesis of SnO2@PMMA and TiO2@PMMA core–shell nanobeads designed for peptide/protein enrichment and MALDI-TOF MS analysis. Angew Chem Int Ed 2008;47:4204–7. [58] Shen W, Shen C, Xiong H, Lu H, Yang P. A robust new strategy for high-molecular-weight proteome research: a 2-hydroxyethyl agarose/polyacrylamide gel enhanced separation and ZnO–PMMA nanobeads assisted identification. Talanta 2010;82:1594–8. [59] Shen W, Xiong H, Xu Y, Cai S, Lu H, Yang P. ZnO-poly(methyl methacrylate) nanobeads for enriching and desalting low-abundant proteins followed by directly MALDI-TOF MS analysis. Anal Chem 2008;80:6758–63. [60] Chen H, Deng C, Zhang X. Synthesis of Fe3O4@SiO2@PMMA core–shell–shell magnetic microspheres for highly efficient enrichment of peptides and proteins for MALDI-ToF MS analysis. Angew Chem Int Ed 2010;49:607–11. [61] Wong VN, Fernando G, Wagner AR, Zhang J, Kinsel GR, Zauscher S, et al. Separation of peptides with polyionic nanosponges for MALDI-MS analysis. Langmuir 2009;25:1459–65. [62] Mitrovic B, Eastwood S, Wong V, Dyer D, Kinsel G, Scott C. Peptide/protein separation with cationic polymer brush nanosponges for MALDI-MS analysis. Langmuir 2013;29:696–700. [63] Bodenmiller B, Wanka S, Kraft C, Urban J, Campbell D, Pedrioli PG, et al. Phosphoproteomic analysis reveals interconnected system-wide responses to perturbations of kinases and phosphatases in yeast. Sci Signal 2010;3:rs4-rs. [64] Bhalla US, Ram PT, Iyengar R. MAP kinase phosphatase as a locus of flexibility in a mitogen-activated protein kinase signaling network. Science 2002;297:1018–23. [65] Miller ML, Jensen LJ, Diella F, Jørgensen C, Tinti M, Li L, et al. Linear motif atlas for phosphorylationdependent signaling. Sci Signal 2008;1:ra2-ra. [66] Draz H, Goldberg AA, Titorenko VI, Tomlinson Guns ES, Safe SH, Sanderson JT. Diindolylmethane and its halogenated derivatives induce protective autophagy in human prostate cancer cells via induction of the oncogenic protein AEG-1 and activation of AMP-activated protein kinase (AMPK). Cell Signal 2017;40:172–82. [67] Xu X, Deng C, Gao M, Yu W, Yang P, Zhang X. Synthesis of magnetic microspheres with immobilized metal ions for enrichment and direct determination of phosphopeptides by matrix-assisted laser desorption ionization mass spectrometry. Adv Mater 2006;18:3289–93. [68] Tan F, Zhang Y, Mi W, Wang J, Wei J, Cai Y, et al. Enrichment of phosphopeptides by Fe3+-immobilized magnetic nanoparticles for phosphoproteome analysis of the plasma membrane of mouse liver. J Proteome Res 2008;7:1078–87. [69] Ficarro SB, Adelmant G, Tomar MN, Zhang Y, Cheng VJ, Marto JA. Magnetic bead processor for rapid evaluation and optimization of parameters for phosphopeptide enrichment. Anal Chem 2009;81:4566–75. [70] Li Y-C, Lin Y-S, Tsai P-J, Chen C-T, Chen W-Y, Chen Y-C. Nitrilotriacetic acid-coated magnetic nanoparticles as affinity probes for enrichment of histidine-tagged proteins and phosphorylated peptides. Anal Chem 2007;79:7519–25. [71] Ma W, Zhang Y, Li L, Zhang Y, Yu M, Guo J, et al. Ti4+-immobilized magnetic composite microspheres for highly selective enrichment of phosphopeptides. Adv Funct Mater 2013;23:107–15. [72] Zhang L, Zhao Q, Liang Z, Yang K, Sun L, Zhang L, et al. Synthesis of adenosine functionalized metal immobilized magnetic nanoparticles for highly selective and sensitive enrichment of phosphopeptides. Chem Commun 2012;48:6274–6. [73] Yan Y, Zheng Z, Deng C, Zhang X, Yang P. Facile synthesis of Ti4+-immobilized Fe3O4@polydopamine core-shell microspheres for highly selective enrichment of phosphopeptides. Chem Commun 2013;49:5055–7.

REFERENCES

77

[74] Xu D, Gao M, Deng C, Zhang X. Ultrasensitive enrichment of phosphopeptides with Ti4+ immobilized SiO2 graphene-like multilayer nanosheets. Analyst 2016;141:3421–7. [75] Wei J, Zhang Y, Wang J, Tan F, Liu J, Cai Y, et al. Highly efficient enrichment of phosphopeptides by magnetic nanoparticles coated with zirconium phosphonate for phosphoproteome analysis. Rapid Commun Mass Spectrom 2008;22:1069–80. [76] Qi D, Mao Y, Lu J, Deng C, Zhang X. Phosphate-functionalized magnetic microspheres for immobilization of Zr4+ ions for selective enrichment of the phosphopeptides. J Chromatogr A 2010;1217:2606–17. [77] Li X-S, Wu J-H, Zhao Y, Zhang W-P, Gao Q, Guo L, et al. Preparation of magnetic polymer material with phosphate group and its application to the enrichment of phosphopeptides. J Chromatogr A 2011;1218: 3845–53. [78] Li X-S, Xu L-D, Zhu G-T, Yuan B-F, Feng Y-Q. Zirconium arsenate-modified magnetic nanoparticles: preparation, characterization and application to the enrichment of phosphopeptides. Analyst 2012;137: 959–67. [79] Hwang L, Ayaz-Guner S, Gregorich ZR, Cai W, Valeja SG, Jin S, et al. Specific enrichment of phosphoproteins using functionalized multivalent nanoparticles. J Am Chem Soc 2015;137:2432–5. [80] Chen B, Hwang L, Ochowicz W, Lin Z, Guardado-Alvarez TM, Cai W, et al. Coupling functionalized cobalt ferrite nanoparticle enrichment with online LC/MS/MS for top-down phosphoproteomics. Chem Sci 2017;8:4306–11. [81] Li Y, Qi D, Deng C, Yang P, Zhang X. Cerium ion-chelated magnetic silica microspheres for enrichment and direct determination of phosphopeptides by matrix-assisted laser desorption ionization mass spectrometry. J Proteome Res 2008;7:1767–77. [82] Wang Z-G, Cheng G, Liu Y-L, Zhang J-L, Sun D-H, Ni J-Z. Novel core–shell Cerium(IV)-immobilized magnetic polymeric microspheres for selective enrichment and rapid separation of phosphopeptides. J Colloid Interface Sci 2014;417:217–26. [83] Mirza MR, Rainer M, Messner CB, Guzel Y, Schemeth D, Stasyk T, et al. A new type of metal chelate affinity chromatography using trivalent lanthanide ions for phosphopeptide enrichment. Analyst 2013;138: 2995–3004. [84] Zhao P-X, Guo X-F, Wang H, Qi C-B, Xia H-S, Zhang H-S. Zirconium arsenate-modified silica nanoparticles for specific capture of phosphopeptides and direct analysis by matrix-assisted laser desorption/ionization mass spectrometry. Anal Bioanal Chem 2012;402:1041–56. [85] Zhou H, Xu S, Ye M, Feng S, Pan C, Jiang X, et al. Zirconium phosphonate-modified porous silicon for highly specific capture of phosphopeptides and MALDI-TOF MS analysis. J Proteome Res 2006;5:2431–7. [86] Zhao L, Qin H, Hu Z, Zhang Y, Wu RA, Zou H. A poly(ethylene glycol)-brush decorated magnetic polymer for highly specific enrichment of phosphopeptides. Chem Sci 2012;3:2828–38. [87] Xiong Z, Zhang L, Fang C, Zhang Q, Ji Y, Zhang Z, et al. Ti4+-immobilized multilayer polysaccharide coated magnetic nanoparticles for highly selective enrichment of phosphopeptides. J Mater Chem B 2014;2: 4473–80. [88] Dunn JD, Reid GE, Bruening ML. Techniques for phosphopeptide enrichment prior to analysis by mass spectrometry. Mass Spectrom Rev 2010;29:29–54. [89] Larsen MR, Thingholm TE, Jensen ON, Roepstorff P, Jorgensen TJD. Highly selective enrichment of phosphorylated peptides from peptide mixtures using titanium dioxide microcolumns. Mol Cell Proteomics 2005;4:873–86. [90] Kweon HK, Hakansson K. Selective zirconium dioxide-based enrichment of phosphorylated peptides for mass spectrometric analysis. Anal Chem 2006;78:1743–9. [91] Cheng G, Zhang J-L, Liu Y-L, Sun D-H, Ni J-Z. Synthesis of novel Fe3O4@SiO2@CeO2 microspheres with mesoporous shell for phosphopeptide capturing and labeling. Chem Commun 2011;47:5732–4. [92] Hasan N, Wu H-F. Highly selective and sensitive enrichment of phosphopeptides via NiO nanoparticles using a microwave-assisted centrifugation on-particle ionization/enrichment approach in MALDI-MS. Anal Bioanal Chem 2011;400:3451–62.

78

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

[93] Han L, Shan Z, Chen D, Yu X, Yang P, Tu B, et al. Mesoporous Fe2O3 microspheres: rapid and effective enrichment of phosphopeptides for MALDI-TOF MS analysis. J Colloid Interface Sci 2008;318:315–21. [94] Rivera JG, Choi YS, Vujcic S, Wood TD, Colon LA. Enrichment/isolation of phosphorylated peptides on hafnium oxide prior to mass spectrometric analysis. Analyst 2009;134:31–3. [95] Li Y, Liu Y, Tang J, Lin H, Yao N, Shen X, et al. Fe3O4@Al2O3 magnetic core-shell microspheres for rapid and highly specific capture of phosphopeptides with mass spectrometry analysis. J Chromatogr A 2007;1172:57–71. [96] Li Y, Lin H, Deng C, Yang P, Zhang X. Highly selective and rapid enrichment of phosphorylated peptides using gallium oxide-coated magnetic microspheres for MALDI-TOF-MS and nano-LC-ESI-MS/MS/MS analysis. Proteomics 2008;8:238–49. [97] Chen W-Y, Chen Y-C. Functional Fe3O4@ZnO magnetic nanoparticle-assisted enrichment and enzymatic digestion of phosphoproteins from saliva. Anal Bioanal Chem 2010;398:2049–57. [98] Sturm M, Leitner A, Smatt J-H, Linden M, Lindner W. Tin dioxide microspheres as a promising material for phosphopeptide enrichment prior to liquid chromatography-(tandem) mass spectrometry analysis. Adv Funct Mater 2008;18:2381–9. [99] Jabeen F, Hussain D, Fatima B, Musharraf SG, Huck CW, Bonn GK, et al. Silica-lanthanum oxide: pioneer composite of rare-earth metal oxide in selective phosphopeptides enrichment. Anal Chem 2012; 84:10180–5. [100] Qi D, Lu J, Deng C, Zhang X. Development of core–shell structure Fe3O4@Ta2O5 microspheres for selective enrichment of phosphopeptides for mass spectrometry analysis. J Chromatogr A 2009;1216:5533–9. [101] Tan F, Zhang Y, Wang J, Wei J, Cai Y, Qian X. An efficient method for dephosphorylation of phosphopeptides by cerium oxide. J Mass Spectrom 2008;43:628–32. [102] Zhong H, Xiao X, Zheng S, Zhang W, Ding M, Jiang H, et al. Mass spectrometric analysis of mono- and multi-phosphopeptides by selective binding with NiZnFe2O4 magnetic nanoparticles. Nat Commun 2013;4:. [103] Wu J-H, Li X-S, Zhao Y, Gao Q, Guo L, Feng Y-Q. Titania coated magnetic mesoporous hollow silica microspheres: fabrication and application to selective enrichment of phosphopeptides. Chem Commun 2010;46:9031–3. [104] Ma W-F, Zhang Y, Li L-L, You L-J, Zhang P, Zhang Y-T, et al. Tailor-made magnetic Fe3O4@mTiO2 microspheres with a tunable mesoporous anatase shell for highly selective and effective enrichment of phosphopeptides. ACS Nano 2012;6:3179–88. [105] Zhang Y, Li L, Ma W, Zhang Y, Yu M, Guo J, et al. Two-in-one strategy for effective enrichment of phosphopeptides using magnetic mesoporous γ-Fe2O3 nanocrystal clusters. ACS Appl Mater Interfaces 2013;5:614–21. [106] Li Y, Wu J, Qi D, Xu X, Deng C, Yang P, et al. Novel approach for the synthesis of Fe3O4@TiO2 core-shell microspheres and their application to the highly specific capture of phosphopeptides for MALDI-TOF MS analysis. Chem Commun 2008;564–6. [107] Li Y, Leng T, Lin H, Deng C, Xu X, Yao N, et al. Preparation of Fe3O4@ZrO2 core–shell microspheres as affinity probes for selective enrichment and direct determination of phosphopeptides using matrix-assisted laser desorption ionization mass spectrometry. J Proteome Res 2007;6:4498–510. [108] Lin H-Y, Chen W-Y, Chen Y-C. Iron oxide/tantalum oxide core–shell magnetic nanoparticle-based microwave-assisted extraction for phosphopeptide enrichment from complex samples for MALDI MS analysis. Anal Bioanal Chem 2009;394:2129–36. [109] Lin H-Y, Chen W-Y, Chen Y-C. Iron oxide/niobium oxide core-shell magnetic nanoparticle-based phosphopeptide enrichment from biological samples for MALDI MS analysis. J Biomed Nanotechnol 2009;5:215–23. [110] Qi D, Lu J, Deng C, Zhang X. Magnetically responsive Fe3O4@C@SnO2 core–shell microspheres: synthesis, characterization and application in phosphoproteomics. J Phys Chem C 2009;113:15854–61.

REFERENCES

79

[111] Cheng G, Wang Z-G, Liu Y-L, Zhang J-L, Sun D-H, Ni J-Z. Magnetic affinity microspheres with meso-/ macroporous shells for selective enrichment and fast separation of phosphorylated biomolecules. ACS Appl Mater Interfaces 2013;5:3182–90. [112] Cheng G, Li S-M, Wang Y, Wang Z-G, Zhang J-L, Ni J-Z. REPO4 (RE ¼ La, Nd, Eu) affinity nanorods modified on a MALDI plate for rapid capture of target peptides from complex biosamples. Chem Commun 2013;49:8492–4. [113] Cheng G, Liu Y-L, Wang Z-G, Li S-M, Zhang J-L, Ni J-Z. Yolk-shell magnetic microspheres with mesoporous yttrium phosphate shells for selective capture and identification of phosphopeptides. J Mater Chem B 2013;1:3661–9. [114] Cheng G, Liu Y-L, Zhang J-L, Sun D-H, Ni J-Z. Lanthanum silicate coated magnetic microspheres as a promising affinity material for phosphopeptide enrichment and identification. Anal Bioanal Chem 2012;404:763–70. [115] Wang Z-G, Cheng G, Liu Y-L, Zhang J-L, Sun D-H, Ni J-Z. Magnetic [gamma]-Fe2O3@REVO4 (RE ¼ Sm, Dy, Ho) affinity microspheres for selective capture, fast separation and easy identification of phosphopeptides. J Mater Chem B 2013;1:1491–500. [116] Wang Z-G, Cheng G, Liu Y-L, Zhang J-L, Sun D-H, Ni J-Z. Novel 3D flowerlike hierarchical [gamma]Fe2O3@xNH4F[middle dot]yLuF3 core-shell microspheres tailor-made by a phase transformation process for the capture of phosphopeptides. J Mater Chem B 2013;1:4845–54. [117] Fang G, Gao W, Deng Q, Qian K, Han H, Wang S. Highly selective capture of phosphopeptides using a nano titanium dioxide–multiwalled carbon nanotube nanocomposite. Anal Biochem 2012;423:210–7. [118] Tang LAL, Wang J, Lim TK, Bi X, Lee WC, Lin Q, et al. High-performance graphene-titania platform for detection of phosphopeptides in cancer cells. Anal Chem 2012;84:6693–700. [119] Min Q, Zhang X, Zhang H, Zhou F, Zhu J-J. Synthesis of Fe3O4-graphene-TiO2 ternary composite networks for enhanced capture of phosphopeptides. Chem Commun 2011;47:11709–11. [120] Yan Y, Sun X, Deng C, Li Y, Zhang X. Metal oxide affinity chromatography platform–polydopamine coupled functional two-dimensional titania graphene nanohybrid for phosphoproteome research. Anal Chem 2014;86:4327–32. [121] Wan H, Yan J, Yu L, Zhang X, Xue X, Li X, et al. Zirconia layer coated mesoporous silica microspheres used for highly specific phosphopeptide enrichment. Talanta 2010;82:1701–7. [122] Qian K, Wan J, Liu F, Girault HH, Liu B, Yu C. A phospho-directed macroporous alumina-silica nanoreactor with multi-functions. ACS Nano 2009;3:3656–62. [123] Yan Y, Zhang X, Deng C. Designed synthesis of titania nanoparticles coated hierarchially ordered macro/ mesoporous silica for selective enrichment of phosphopeptides. ACS Appl Mater Interfaces 2014;6: 5467–71. [124] Zhang Y, Wang H, Lu H. Sequential selective enrichment of phosphopeptides and glycopeptides using amine-functionalized magnetic nanoparticles. Mol BioSyst 2013;9:492–500. [125] Fischnaller M, Bakry R, Vallant RM, Huber LA, Bonn GK. C60-fullerene bound silica for the preconcentration and the fractionation of multiphosphorylated peptides. Anal Chim Acta 2013;761:92–101. [126] Atakay M, Celikbicak O, Salih B. Amine-functionalized sol-gel-based lab-in-a-pipet-tip approach for the fast enrichment and specific purification of phosphopeptides in MALDI-MS applications. Anal Chem 2012;84:2713–20. [127] Chen C-T, Wang L-Y, Ho Y-P. Use of polyethylenimine-modified magnetic nanoparticles for highly specific enrichment of phosphopeptides for mass spectrometric analysis. Anal Bioanal Chem 2011;399: 2795–806. [128] Chang C-K, Wu C-C, Wang Y-S, Chang H-C. Selective extraction and enrichment of multiphosphorylated peptides using polyarginine-coated diamond nanoparticles. Anal Chem 2008;80:3791–7. [129] Xu L-N, Li L-P, Jin L, Bai Y, Liu H-W. Guanidyl-functionalized graphene as a bifunctional adsorbent for selective enrichment of phosphopeptides. Chem Commun 2014;50:10963–6.

80

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

[130] Xiong Z, Chen Y, Zhang L, Ren J, Zhang Q, Ye M, et al. Facile synthesis of guanidyl-functionalized magnetic polymer microspheres for tunable and specific capture of global phosphopeptides or only multiphosphopeptides. ACS Appl Mater Interfaces 2014;6:22743–50. [131] Deng Q, Wu J, Chen Y, Zhang Z, Wang Y, Fang G, et al. Guanidinium functionalized superparamagnetic silica spheres for selective enrichment of phosphopeptides and intact phosphoproteins from complex mixtures. J Mater Chem B 2014;2:1048–58. [132] Saeed A, Maya F, Xiao DJ, Najam-ul-Haq M, Svec F, Britt DK. Growth of a highly porous coordination polymer on a macroporous polymer monolith support for enhanced immobilized metal ion affinity chromatographic enrichment of phosphopeptides. Adv Funct Mater 2014;24:5790–7. [133] Pinho SS, Reis CA. Glycosylation in cancer: mechanisms and clinical implications. Nat Rev Cancer 2015;15:540–55. [134] Gilar M, Yu Y-Q, Ahn J, Xie H, Han H, Ying W, et al. Characterization of glycoprotein digests with hydrophilic interaction chromatography and mass spectrometry. Anal Biochem 2011;417:80–8. [135] Kuo C-W, Wu IL, Hsiao H-H, Khoo K-H. Rapid glycopeptide enrichment and N-glycosylation site mapping strategies based on amine-functionalized magnetic nanoparticles. Anal Bioanal Chem 2012;402:2765–76. [136] Jin T, Xiong Z, Zhu X, Mehio N, Chen Y, Hu J, et al. Template-free synthesis of mesoporous polymers for highly selective enrichment of glycopeptides. ACS Macro Lett 2015;4:570–4. [137] Miao W, Zhang C, Cai Y, Zhang Y, Lu H. Fast solid-phase extraction of N-linked glycopeptides by aminefunctionalized mesoporous silica nanoparticles. Analyst 2016;141:2435–40. [138] Wang Y, Wang J, Gao M, Zhang X. Functional dual hydrophilic dendrimer-modified metal-organic framework for the selective enrichment of N-glycopeptides. Proteomics 2017;17:1700005. [139] Sun X, Yang G, Sun S, Quan R, Dai W, Li B, et al. The hydroxyl-functionalized magnetic particles for purification of glycan-binding proteins. Curr Pharm Biotechnol 2009;10:753–60. [140] Xiong Z, Zhao L, Wang F, Zhu J, Qin H, Wu RA, et al. Synthesis of branched PEG brushes hybrid hydrophilic magnetic nanoparticles for the selective enrichment of N-linked glycopeptides. Chem Commun 2012;48:8138–40. [141] He X-M, Liang X-C, Chen X, Yuan B-F, Zhou P, Zhang L-N, et al. High strength and hydrophilic chitosan microspheres for the selective enrichment of N-glycopeptides. Anal Chem 2017;89:9712–21. [142] Li X, Xiong Y, Qing G, Jiang G, Li X, Sun T, et al. Bioinspired saccharide-saccharide interaction and smart polymer for specific enrichment of sialylated glycopeptides. ACS Appl Mater Interfaces 2016;8:13294–302. [143] Ji Y, Xiong Z, Huang G, Liu J, Zhang Z, Liu Z, et al. Efficient enrichment of glycopeptides using metalorganic frameworks by hydrophilic interaction chromatography. Analyst 2014;139:4987–93. [144] Jiao F, Gao F, Wang H, Deng Y, Zhang Y, Qian X, et al. Ultrathin Au nanowires assisted magnetic graphene-silica ZIC-HILIC composites for highly specific enrichment of N-linked glycopeptides. Anal Chim Acta 2017;970:47–56. [145] Wu R, Li L, Deng C. Highly efficient and selective enrichment of glycopeptides using easily synthesized magG/PDA/Au/L-Cys composites. Proteomics 2016;16:1311–20. [146] Liu J, Yang K, Shao W, Li S, Wu Q, Zhang S, et al. Synthesis of zwitterionic polymer particles via combined distillation precipitation polymerization and click chemistry for highly efficient enrichment of glycopeptide. ACS Appl Mater Interfaces 2016;8:22018–24. [147] Ma W, Xu L, Li X, Shen S, Wu M, Bai Y, et al. Cysteine-functionalized metal-organic framework: facile synthesis and high efficient enrichment of N-linked glycopeptides in cell lysate. ACS Appl Mater Interfaces 2017;9:19562–8. [148] Li J, Liu J, Liu Z, Tan Y, Liu X, Wang F. Detecting proteins glycosylation by a homogeneous reaction system with zwitterionic gold nanoclusters. Anal Chem 2017;89:4339–43. [149] Dong X, Qin H, Mao J, Yu D, Li X, Shen A, et al. In-depth analysis of glycoprotein sialylation in serum using a dual-functional material with superior hydrophilicity and switchable surface charge. Anal Chem 2017;89:3966–72.

REFERENCES

81

[150] Wang Y, Wang J, Gao M, Zhang X. A novel carbon material with nanopores prepared using a metal-organic framework as precursor for highly selective enrichment of N-linked glycans. Anal Bioanal Chem 2017;409:431–8. [151] Huang G, Xiong Z, Qin H, Zhu J, Sun Z, Zhang Y, et al. Synthesis of zwitterionic polymer brushes hybrid silica nanoparticles via controlled polymerization for highly efficient enrichment of glycopeptides. Anal Chim Acta 2014;809:61–8. [152] Chen Y, Xiong Z, Zhang L, Zhao J, Zhang Q, Peng L, et al. Facile synthesis of zwitterionic polymer-coated core-shell magnetic nanoparticles for highly specific capture of N-linked glycopeptides. Nanoscale 2015;7:3100–8. [153] Pan Y, Ma C, Tong W, Fan C, Zhang Q, Zhang W, et al. Preparation of sequence-controlled triblock copolymer-grafted silica microparticles by sequential-ATRP for highly efficient glycopeptides enrichment. Anal Chem 2015;87:656–62. [154] Xie Y, Deng C, Li Y. Designed synthesis of ultra-hydrophilic sulfo-functionalized metal-organic frameworks with a magnetic core for highly efficient enrichment of the N-linked glycopeptides. J Chromatogr A 2017;1508:1–6. [155] Zou X, Jie J, Yang B. Single-step enrichment of N-glycopeptides and phosphopeptides with novel multifunctional Ti4+-immobilized dendritic polyglycerol coated chitosan nanomaterials. Anal Chem 2017;89:7520–6. [156] Palmisano G, Lendal SE, Engholm-Keller K, Leth-Larsen R, Parker BL, Larsen MR. Selective enrichment of sialic acid-containing glycopeptides using titanium dioxide chromatography with analysis by HILIC and mass spectrometry. Nat Protoc 2010;5:1974–82. [157] Ma W-F, Li L-L, Zhang Y, An Q, You L-J, Li J-M, et al. Ligand-free strategy for ultrafast and highly selective enrichment of glycopeptides using Ag-coated magnetic nanoarchitectures. J Mater Chem 2012;22:23981–8. [158] Zhang M, Liu Y, Zhang D, Chen T, Li Z. Facile and selective enrichment of intact sialoglycopeptides using graphitic carbon nitride. Anal Chem 2017;89:8064–9. [159] Wang J, Li J, Wang Y, Gao M, Zhang X, Yang P. Development of versatile metal-organic framework functionalized magnetic graphene core-shell biocomposite for highly specific recognition of glycopeptides. ACS Appl Mater Interfaces 2016;8:27482–9. [160] Zauner G, Deelder AM, Wuhrer M. Recent advances in hydrophilic interaction liquid chromatography (HILIC) for structural glycomics. Electrophoresis 2011;32:3456–66. [161] Feng X, Deng C, Gao M, Zhang X. Facile and easily popularized synthesis of L-cysteine-functionalized magnetic nanoparticles based on one-step functionalization for highly efficient enrichment of glycopeptides. Anal Bioanal Chem 2017;410:989–98. [162] Zhang Y, Zhang C, Jiang H, Yang P, Lu H. Fishing the PTM proteome with chemical approaches using functional solid phases. Chem Soc Rev 2015;44:8260–87. [163] Ongay S, Boichenko A, Govorukhina N, Bischoff R. Glycopeptide enrichment and separation for protein glycosylation analysis. J Sep Sci 2012;35:2341–72. [164] Lin Z-A, Zheng J-N, Lin F, Zhang L, Cai Z, Chen G-N. Synthesis of magnetic nanoparticles with immobilized aminophenylboronic acid for selective capture of glycoproteins. J Mater Chem 2011;21:518–24. [165] Lin Z, Zheng J, Xia Z, Yang H, Zhang L, Chen G. One-pot synthesis of phenylboronic acid-functionalized core-shell magnetic nanoparticles for selective enrichment of glycoproteins. RSC Adv 2012;2:5062–5. [166] Qi D, Zhang H, Tang J, Deng C, Zhang X. Facile synthesis of mercaptophenylboronic acid-functionalized core–shell structure Fe3O4@C@Au magnetic microspheres for selective enrichment of glycopeptides and glycoproteins. J Phys Chem C 2010;114:9221–6. [167] Zhang L, Xu Y, Yao H, Xie L, Yao J, Lu H, et al. Boronic acid functionalized core–satellite composite nanoparticles for advanced enrichment of glycopeptides and glycoproteins. Chem Eur J 2009;15:10158–66. [168] Wang H, Bie Z, Lu C, Liu Z. Magnetic nanoparticles with dendrimer-assisted boronate avidity for the selective enrichment of trace glycoproteins. Chem Sci 2013;4:4298–303.

82

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

[169] Zhang X, Wang J, He X, Chen L, Zhang Y. Tailor-made boronic acid functionalized magnetic nanoparticles with a tunable polymer shell-assisted for the selective enrichment of glycoproteins/glycopeptides. ACS Appl Mater Interfaces 2015;7:24576–84. [170] Xu Y, Wu Z, Zhang L, Lu H, Yang P, Webley PA, et al. Highly specific enrichment of glycopeptides using boronic acid-functionalized mesoporous silica. Anal Chem 2009;81:503–8. [171] Liu L, Yu M, Zhang Y, Wang C, Lu H. Hydrazide functionalized core–shell magnetic nanocomposites for highly specific enrichment of N-glycopeptides. ACS Appl Mater Interfaces 2014;6:7823–32. [172] Yeap WS, Tan YY, Loh KP. Using detonation nanodiamond for the specific capture of glycoproteins. Anal Chem 2008;80:4659–65. [173] Xu G, Zhang W, Wei L, Lu H, Yang P. Boronic acid-functionalized detonation nanodiamond for specific enrichment of glycopeptides in glycoproteome analysis. Analyst 2013;138:1876–85. [174] Xu Y, Zhang L, Lu H, Yang P. On-plate enrichment of glycopeptides by using boronic acid functionalized gold-coated Si wafer. Proteomics 2010;10:1079–86. [175] Zhang H, Li X-j, Martin DB, Aebersold R. Identification and quantification of N-linked glycoproteins using hydrazide chemistry, stable isotope labeling and mass spectrometry. Nat Biotechnol 2003;21:660. [176] Tran TH, Park S, Lee H, Park S, Kim B, Kim O-H, et al. Ultrasmall gold nanoparticles for highly specific isolation/enrichment of N-linked glycosylated peptides. Analyst 2012;137:991–8. [177] Zou Z, Ibisate M, Zhou Y, Aebersold R, Xia Y, Zhang H. Synthesis and evaluation of superparamagnetic silica particles for extraction of glycopeptides in the microtiter plate format. Anal Chem 2008;80:1228–34. [178] Sun S, Yang G, Wang T, Wang Q, Chen C, Li Z. Isolation of N-linked glycopeptides by hydrazinefunctionalized magnetic particles. Anal Bioanal Chem 2010;396:3071–8. [179] Lis H, Sharon N, Katchalski E. Identification of the carbohydrate-protein linking group in soybean hemagglutinin. Biochim Biophys Acta Gen Subj 1969;192:364–6. [180] Fanayan S, Hincapie M, Hancock WS. Using lectins to harvest the plasma/serum glycoproteome. Electrophoresis 2012;33:1746–54. [181] Tang J, Liu Y, Yin P, Yao G, Yan G, Deng C, et al. Concanavalin A-immobilized magnetic nanoparticles for selective enrichment of glycoproteins and application to glycoproteomics in hepatocelluar carcinoma cell line. Proteomics 2010;10:2000–14. [182] Liu Y, Fu D, Yu L, Xiao Y, Peng X, Liang X. Oxidized dextran facilitated synthesis of a silica-based concanavalin a material for lectin affinity enrichment of glycoproteins/glycopeptides. J Chromatogr A 2016;1455:147–55. [183] Calvano CD, Zambonin CG, Jensen ON. Assessment of lectin and HILIC based enrichment protocols for characterization of serum glycoproteins by mass spectrometry. J Proteome 2008;71:304–17. [184] Angel PM, Mehta A, Norris-Caneda K, Drake RR. MALDI imaging mass spectrometry of N-glycans and tryptic peptides from the same formalin-fixed, paraffin-embedded tissue section. Totowa, NJ: Humana Press. p. 1-17. [185] Safdar M, Sproß J, J€anis J. Microscale immobilized enzyme reactors in proteomics: Latest developments. J Chromatogr A 2014;1324:1–10. [186] Li Y, Zhang X, Deng C. Functionalized magnetic nanoparticles for sample preparation in proteomics and peptidomics analysis. Chem Soc Rev 2013;42:8517–39. [187] Le Nel A, Krenkova J, Kleparnik K, Smadja C, Taverna M, Viovy J-L, et al. On-chip tryptic digest with direct coupling to ESI-MS using magnetic particles. Electrophoresis 2008;29:4944–7. [188] Lin S, Yao G, Qi D, Li Y, Deng C, Yang P, et al. Fast and efficient proteolysis by microwave-assisted protein digestion using trypsin-immobilized magnetic silica microspheres. Anal Chem 2008;80:3655–65. [189] Lin S, Yun D, Qi D, Deng C, Li Y, Zhang X. Novel microwave-assisted digestion by trypsin-immobilized magnetic nanoparticles for proteomic analysis. J Proteome Res 2008;7:1297–307. [190] Sustrova B, Novotna L, Kucerova Z, Ticha M. Immobilization of α-chymotrypsin to magnetic particles and their use for proteolytic cleavage of porcine pepsin A. J Mol Catal B Enzym 2009;60:22–8.

REFERENCES

83

[191] Li D, Teoh WY, Gooding JJ, Selomulya C, Amal R. Functionalization strategies for protease immobilization on magnetic nanoparticles. Adv Funct Mater 2010;20:1767–77. [192] Liu J, Lin S, Qi D, Deng C, Yang P, Zhang X. On-chip enzymatic microreactor using trypsin-immobilized superparamagnetic nanoparticles for highly efficient proteolysis. J Chromatogr A 2007;1176:169–77. [193] Li Y, Wojcik R, Dovichi NJ. A replaceable microreactor for on-line protein digestion in a two-dimensional capillary electrophoresis system with tandem mass spectrometry detection. J Chromatogr A 2011;1218: 2007–11. [194] Qin W, Song Z, Fan C, Zhang W, Cai Y, Zhang Y, et al. Trypsin immobilization on hairy polymer chains hybrid magnetic nanoparticles for ultra fast, highly efficient proteome digestion, facile 18O labeling and absolute protein quantification. Anal Chem 2012;84:3138–44. [195] Li Y, Xu X, Yan B, Deng C, Yu W, Yang P, et al. Microchip reactor packed with metal-ion chelated magnetic silica microspheres for highly efficient proteolysis. J Proteome Res 2007;6:2367–75. [196] Li Y, Yan B, Xu X, Deng C, Yang P, Shen X, et al. On-column tryptic mapping of proteins using metal-ionchelated magnetic silica microspheres by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 2007;21:2263–8. [197] Fan J, Shui WQ, Yang PY, Wang XY, Xu YM, Wang HH, et al. Mesoporous silica nanoreactors for highly efficient proteolysis. Chem Eur J 2005;11:5391–6. [198] Schlossbauer A, Schaffert D, Kecht J, Wagner E, Bein T. Click chemistry for high-density biofunctionalization of mesoporous silica. J Am Chem Soc 2008;130:12558. [199] Min Q, Wu RA, Zhao L, Qin H, Ye M, Zhu J-J, et al. Size-selective proteolysis on mesoporous silica-based trypsin nanoreactor for low-MW proteome analysis. Chem Commun 2010;46:6144–6. [200] Zhang Y, Cao W, Liu M, Yang S, Wu H, Lu H, et al. Immobilization of trypsin on water-soluble dendrimermodified carbon nanotubes for on-plate proteolysis combined with MALDI-MS analysis. Mol BioSyst 2010;6:1447–9. [201] Wang S, Bao H, Yang P, Chen G. Immobilization of trypsin in polyaniline-coated nano-Fe3O4/carbon nanotube composite for protein digestion. Anal Chim Acta 2008;612:182–9. [202] Wei L, Zhang W, Lu H, Yang P. Immobilization of enzyme on detonation nanodiamond for highly efficient proteolysis. Talanta 2010;80:1298–304. [203] Jiang B, Yang K, Zhao Q, Wu Q, Liang Z, Zhang L, et al. Hydrophilic immobilized trypsin reactor with magnetic graphene oxide as support for high efficient proteome digestion. J Chromatogr A 2012;1254:8–13. [204] Xu G, Chen X, Hu J, Yang P, Yang D, Wei L. Immobilization of trypsin on graphene oxide for microwaveassisted on-plate proteolysis combined with MALDI-MS analysis. Analyst 2012;137:2757–61. [205] Bao H, Zhang L, Chen G. Immobilization of trypsin via graphene oxide-silica composite for efficient microchip proteolysis. J Chromatogr A 2013;1310:74–81. [206] Shih Y-H, Lo S-H, Yang N-S, Singco B, Cheng Y-J, Wu C-Y, et al. Trypsin-immobilized metal–organic framework as a biocatalyst in proteomics analysis. ChemPlusChem 2012;77:982–6. [207] Lu M, Yang X, Yang Y, Qin P, Wu X, Cai Z. Nanomaterials as assisted matrix of laser desorption/ionization time-of-flight mass spectrometry for the analysis of small molecules. Nanomaterials 2017;7:87. [208] Xu SY, Li YF, Zou HF, Qiu JS, Guo Z, Guo BC. Carbon nanotubes as assisted matrix for laser desorption/ ionization time-of-flight mass spectrometry. Anal Chem 2003;75:6191–5. [209] Ugarov MV, Egan T, Khabashesku DV, Schultz JA, Peng H, Khabashesku VN, et al. MALDI matrices for biomolecular analysis based on functionalized carbon nanomaterials. Anal Chem 2004;76:6734–42. [210] Ren SF, Guo YL. Oxidized carbon nanotubes as matrix for matrix-assisted laser desorption/ionization timeof-flight mass spectrometric analysis of biomolecules. Rapid Commun Mass Spectrom 2005;19:255–60. [211] Pan C, Xu S, Zou H, Guo Z, Zhang Y, Guo B. Carbon nanotubes as adsorbent of solid-phase extraction and matrix for laser desorption/ionization mass spectrometry. J Am Soc Mass Spectrom 2005;16:263–70. [212] Wu C-L, Wang C-C, Lai Y-H, Lee H, Lin J-D, Lee YT, et al. Selective enhancement of carbohydrate ion abundances by diamond nanoparticles for mass spectrometric analysis. Anal Chem 2013;85:3836–41.

84

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

[213] Coffinier Y, Szunerits S, Drobecq H, Melnyk O, Boukherroub R. Diamond nanowires for highly sensitive matrix-free mass spectrometry analysis of small molecules. Nanoscale 2012;4:231–8. [214] Dong X, Cheng J, Li J, Wang Y. Graphene as a novel matrix for the analysis of small molecules by MALDITOF MS. Anal Chem 2010;82:6208–14. [215] Kim Y-K, Na H-K, Kwack S-J, Ryoo S-R, Lee Y, Hong S, et al. Synergistic effect of graphene oxide/ MWCNT films in laser desorption/ionization mass spectrometry of small molecules and tissue imaging. ACS Nano 2011;5:4550–61. [216] Abdelhamid HN, Wu B-S, Wu H-F. Graphene coated silica applied for high ionization matrix assisted laser desorption/ionization mass spectrometry: a novel approach for environmental and biomolecule analysis. Talanta 2014;126:27–37. [217] Lee G, Bae S-E, Huh S, Cha S. Graphene oxide embedded sol-gel (GOSG) film as a SALDI MS substrate for robust metabolite fingerprinting. RSC Adv 2015;5:56455–9. [218] Hinman SS, Chen C-Y, Duan J, Cheng Q. Calcinated gold nanoparticle arrays for on-chip, multiplexed and matrix-free mass spectrometric analysis of peptides and small molecules. Nanoscale 2016;8:1665–75. [219] Silina YE, Meier F, Nebolsin VA, Koch M, Volmer DA. Novel galvanic nanostructures of Ag and Pd for efficient laser desorption/ionization of low molecular weight compounds. J Am Soc Mass Spectrom 2014;25:841–51. [220] Park EJ, Han SW, Jeong B, Park SH, Kim Y-G, Kim YH, et al. Effect of polydimethylsiloxane (PDMS) coating on TiO2-based MALDI matrix for dimethyl methylphosphonate (DMMP) analysis. Appl Surf Sci 2015;353:342–9. [221] Shin WJ, Shin JH, Song JY, Han SY. Effects of ZnO nanowire length on surface-assisted laser desorption/ ionization of small molecules. J Am Soc Mass Spectrom 2010;21:989–92. [222] Kawasaki H, Yonezawa T, Watanabe T, Arakawa R. Platinum nanoflowers for surface-assisted laser desorption/ionization mass spectrometry of biomolecules. J Phys Chem C 2007;111:16278–83. [223] Zhang X, Wang F, Xia Y. Trypsin functionalization and zirconia coating of mesoporous silica nanotubes for matrix-assisted laser desorption/ionization mass spectrometry analysis of phosphoprotein. J Chromatogr A 2013;1306:20–6. [224] Kailasa SK, Wu H-F. Surface modified BaTiO3 nanoparticles as the matrix for phospholipids and as extracting probes for LLME of hydrophobic proteins in Escherichia coli by MALDI–MS. Talanta 2013;114: 283–90. [225] McLean JA, Stumpo KA, Russell DH. Size-selected (2–10 nm) gold nanoparticles for matrix assisted laser desorption ionization of peptides. J Am Chem Soc 2005;127:5304–5. [226] Popovic I, Nesic M, Vranjes M, Saponjic Z, Petkovic M. TiO2 nanocrystals-assisted laser desorption and ionization time-of-flight mass spectrometric analysis of steroid hormones, amino acids and saccharides. Validation and comparison of methods. RSC Adv 2016;6:1027–36. [227] Piret G, Kim D, Drobecq H, Coffinier Y, Melnyk O, Schmuki P, et al. Surface-assisted laser desorptionionization mass spectrometry on titanium dioxide (TiO2) nanotube layers. Analyst 2012;137:3058–63. [228] Kim J-I, Ryu S-Y, Park J-M, Noh J-Y, Kang M-J, Kwak S-Y, et al. Nylon nanoweb with TiO2 nanoparticles as a solid matrix for matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 2014;28:2427–36. [229] Shih Y-H, Chien C-H, Singco B, Hsu C-L, Lin C-H, Huang H-Y. Metal-organic frameworks: new matrices for surface-assisted laser desorption-ionization mass spectrometry. Chem Commun 2013;49:4929–31. [230] Fu C-P, Lirio S, Liu W-L, Lin C-H, Huang H-Y. A novel type of matrix for surface-assisted laser desorption–ionization mass spectrometric detection of biomolecules using metal-organic frameworks. Anal Chim Acta 2015;888:103–9. [231] Lin Z, Bian W, Zheng J, Cai Z. Magnetic metal-organic framework nanocomposites for enrichment and direct detection of small molecules by negative-ion matrix-assisted laser desorption/ionization time-offlight mass spectrometry. Chem Commun 2015;51:8785–8.

REFERENCES

85

[232] Neiswinger J, Uzoma I, Cox E, Rho H, Song G, Paul C, et al. Protein microarrays: flexible tools for scientific innovation. Cold Spring Harb Protoc 2016;2016:pdb.top081471. [233] Nimse SB, Sonawane MD, Song K-S, Kim T. Biomarker detection technologies and future directions. Analyst 2016;141:740–55. [234] Gao W, Wang W, Yao S, Wu S, Zhang H, Zhang J, et al. Highly sensitive detection of multiple tumor markers for lung cancer using gold nanoparticle probes and microarrays. Anal Chim Acta 2017;958:77–84. [235] Gupta S, Huda S, Kilpatrick PK, Velev OD. Characterization and optimization of gold nanoparticle-based silver-enhanced immunoassays. Anal Chem 2007;79:3810–20. [236] Scott AW, Garimella V, Calabrese CM, Mirkin CA. Universal biotin-PEG-linked gold nanoparticle probes for the simultaneous detection of nucleic acids and proteins. Bioconjug Chem 2017;28:203–11. [237] Gao J, Liu D, Wang Z. Screening lectin-binding specificity of bacterium by lectin microarray with gold nanoparticle probes. Anal Chem 2010;82:9240–7. [238] Stevenson R, Ingram A, Leung H, McMillan DC, Graham D. Quantitative SERRS immunoassay for the detection of human PSA. Analyst 2009;134:842–4. [239] Han XX, Kitahama Y, Itoh T, Wang CX, Zhao B, Ozaki Y. Protein-mediated sandwich strategy for surfaceenhanced Raman scattering: application to versatile protein detection. Anal Chem 2009;81:3350–5. [240] Yoon KJ, Seo HK, Hwang H, Pyo D, Eom I-Y, Hahn JH, et al. Bioanalytical application of SERS immunoassay for detection of prostate-specific antigen. Bull Kor Chem Soc 2010;31:1215–8. [241] Li T, Liu D, Wang Z. Microarray-based Raman spectroscopic assay for kinase inhibition by gold nanoparticle probes. Biosens Bioelectron 2009;24:3335–9. [242] Karlin-Neumann G, Sedova M, Falkowski M, Wang Z, Lin S, Jain M. Application of quantum dots to multicolor microarray experiments: four-color genotyping. Methods Molr Biol (Clifton, NJ) 2007;374: 239–51. [243] Sukhanova A, Devy M, Venteo L, Kaplan H, Artemyev M, Oleinikov V, et al. Biocompatible fluorescent nanocrystals for immunolabeling of membrane proteins and cells. Anal Biochem 2004;324:60–7. [244] Sun Y-P, Zhou B, Lin Y, Wang W, Fernando KAS, Pathak P, et al. Quantum-sized carbon dots for bright and colorful photoluminescence. J Am Chem Soc 2006;128:7756–7. [245] Jokerst JV, Raamanathan A, Christodoulides N, Floriano PN, Pollard AA, Simmons GW, et al. Nano-biochips for high performance multiplexed protein detection: determinations of cancer biomarkers in serum and saliva using quantum dot bioconjugate labels. Biosens Bioelectron 2009;24:3622–9. [246] Zajac A, Song D, Qian W, Zhukov T. Protein microarrays and quantum dot probes for early cancer detection. Colloids Surf B: Biointerfaces 2007;58:309–14. [247] Ghazani AA, Lee JA, Klostranec J, Xiang Q, Dacosta RS, Wilson BC, et al. High throughput quantification of protein expression of cancer antigens in tissue microarray using quantum dot nanocrystals. Nano Lett 2006;6:2881–6. [248] Resch-Genger U, Grabolle M, Cavaliere-Jaricot S, Nitschke R, Nann T. Quantum dots versus organic dyes as fluorescent labels. Nat Methods 2008;5:763. [249] Morales-Narvaez E, Monton H, Fomicheva A, Merkoci A. Signal enhancement in antibody microarrays using quantum dots nanocrystals: application to potential Alzheimer’s disease biomarker screening. Anal Chem 2012;84:6821–7. [250] Geho D, Lahar N, Gurnani P, Huebschman M, Herrmann P, Espina V, et al. Pegylated, steptavidinconjugated quantum dots are effective detection elements for reverse-phase protein microarrays. Bioconjug Chem 2005;16:559–66. [251] Liu L, Wu S, Jing F, Zhou H, Jia C, Li G, et al. Bead-based microarray immunoassay for lung cancer biomarkers using quantum dots as labels. Biosens Bioelectron 2016;80:300–6. [252] Bilan R, Ametzazurra A, Brazhnik K, Escorza S, Fernandez D, Uribarri M, et al. Quantum-dot-based suspension microarray for multiplex detection of lung cancer markers: preclinical validation and comparison with the Luminex xMAP (R) system. Sci Rep 2017;7:44668.

86

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

[253] Li Z, Zhang Q, Huang H, Ren C, Pan Y, Wang Q, et al. RGDS-conjugated CdSeTe/CdS quantum dots as near-infrared fluorescent probe: preparation, characterization and bioapplication. J Nanopart Res 2016;18:3456. [254] Yeom SY, Son CH, Kim BS, Tag SH, Nam E, Shin H, et al. Multiplexed detection of epigenetic markers using quantum dot (QD)-encoded hydrogel microparticles. Anal Chem 2016;88:4259–68. [255] Garcia-Cortes M, Ruiz Encinar J, Costa-Fernandez JM, Sanz-Medel A. Highly sensitive nanoparticle-based immunoassays with elemental detection: application to prostate-specific antigen quantification. Biosens Bioelectron 2016;85:128–34. [256] Zheng J, Bai J, Zhou Q, Li J, Li Y, Yang J, et al. DNA-templated in situ growth of AgNPs on SWNTs: a new approach for highly sensitive SERS assay of microRNA. Chem Commun 2015;51:6552–5. [257] Chen Z, Tabakman SM, Goodwin AP, Kattah MG, Daranciang D, Wang X, et al. Protein microarrays with carbon nanotubes as multicolor Raman labels. Nat Biotechnol 2008;26:1285. [258] Landry MP, Ando H, Chen AY, Cao J, Kottadiel VI, Chio L, et al. Single-molecule detection of protein efflux from microorganisms using fluorescent single-walled carbon nanotube sensor arrays. Nat Nanotechnol 2017;12:368–77. [259] Yang C, Denno ME, Pyakurel P, Venton BJ. Recent trends in carbon nanomaterial-based electrochemical sensors for biomolecules: a review. Anal Chim Acta 2015;887:17–37. [260] Nam JM, Thaxton CS, Mirkin CA. Nanoparticle-based bio-bar codes for the ultrasensitive detection of proteins. Science 2003;301:1884–6. [261] Thaxton CS, Elghanian R, Thomas AD, Stoeva SI, Lee J-S, Smith ND, et al. Nanoparticle-based bio-barcode assay redefines “undetectable” PSA and biochemical recurrence after radical prostatectomy. Proc Natl Acad Sci U S A 2009;106:18437–42. [262] Liu H, Wang Y, Li H, Wang Z, Xu D. Luminescent Rhodamine B doped core-shell silica nanoparticle labels for protein microarray detection. Dyes Pigments 2013;98:119–24. [263] Wang X, Matei E, Deng L, Koharudin L, Gronenborn AM, Ramstr€ om O, et al. Sensing lectin–glycan interactions using lectin super-microarrays and glycans labeled with dye-doped silica nanoparticles. Biosens Bioelectron 2013;47:258–64. [264] Giesen C, Mairinger T, Khoury L, Waentig L, Jakubowski N, Panne U. Multiplexed immunohistochemical detection of tumor markers in breast cancer tissue using laser ablation inductively coupled plasma mass spectrometry. Anal Chem 2011;83:8177–83. [265] Yan X, Xu M, Yang L, Wang Q. Absolute quantification of intact proteins via 1,4,7,10-tetraazacyclododecane-1,4,7-trisacetic acid–10-maleimidoethylacetamide–europium labeling and HPLC coupled with species-unspecific isotope dilution ICPMS. Anal Chem 2010;82:1261–9. [266] Konz T, An˜o´n Alvarez E, Montes-Bayon M, Sanz-Medel A. Antibody labeling and elemental mass spectrometry (inductively coupled plasma-mass spectrometry) using isotope dilution for highly sensitive ferritin determination and iron-ferritin ratio measurements. Anal Chem 2013;85:8334–40. [267] Li Y, Sun S-K, J-l Y, Jiang Y. Label-free DNA hybridization detection and single base-mismatch discrimination using CE-ICP-MS assay. Analyst 2011;136:5038–45. [268] Chen P, Wu P, Chen J, Yang P, Zhang X, Zheng C, et al. Label-free and separation-free atomic fluorescence spectrometry based bioassay: sensitive determination of single-strand DNA, protein, and double-strand DNA. Anal Chem 2016;88:2065–71. [269] Hsu IH, Chen W-H, Wu T-K, Sun Y-C. Gold nanoparticle-based inductively coupled plasma mass spectrometry amplification and magnetic separation for the sensitive detection of a virus-specific RNA sequence. J Chromatogr A 2011;1218:1795–801. [270] Zhang S, Liu R, Xing Z, Zhang S, Zhang X. Multiplex miRNA assay using lanthanide-tagged probes and the duplex-specific nuclease amplification strategy. Chem Commun 2016;52:14310–3.

REFERENCES

87

[271] Yang W, Xi Z, Zeng X, Fang L, Jiang W, Wu Y, et al. Magnetic bead-based AuNP labelling combined with inductively coupled plasma mass spectrometry for sensitively and specifically counting cancer cells. J Anal At Spectrom 2016;31:679–85. [272] Zhang Y, Chen B, He M, Yang B, Zhang J, Hu B. Immunomagnetic separation combined with inductively coupled plasma mass spectrometry for the detection of tumor cells using gold nanoparticle labeling. Anal Chem 2014;86:8082–9. [273] Li F, Zhao Q, Wang C, Lu X, Li X-F, Le XC. Detection of Escherichia coli O157:H7 using gold nanoparticle labeling and inductively coupled plasma mass spectrometry. Anal Chem 2010;82:3399–403. [274] Lin Y, Hamme Ii AT. Gold nanoparticle labeling based ICP-MS detection/measurement of bacteria, and their quantitative photothermal destruction. J Mater Chem B 2015;3:3573–82. [275] Zhang C, Wu F, Zhang Y, Wang X, Zhang X. A novel combination of immunoreaction and ICP-MS as a hyphenated technique for the determination of thyroid-stimulating hormone (TSH) in human serum. J Anal At Spectrom 2001;16:1393–6. [276] Zhang C, Zhang Z, Yu B, Shi J, Zhang X. Application of the biological conjugate between antibody and colloid Au nanoparticles as analyte to inductively coupled plasma mass spectrometry. Anal Chem 2002;74:96–9. [277] Baranov VI, Quinn Z, Bandura DR, Tanner SD. A sensitive and quantitative element-tagged immunoassay with ICPMS detection. Anal Chem 2002;74:1629–36. [278] Zhao Q, Lu X, Yuan C-G, Li X-F, Le XC. Aptamer-linked assay for thrombin using gold nanoparticle amplification and inductively coupled plasma–mass spectrometry detection. Anal Chem 2009;81:7484–9. [279] Liu J-M, Li Y, Jiang Y, Yan X-P. Gold nanoparticles amplified ultrasensitive quantification of human urinary protein by capillary electrophoresis with on-line inductively coupled plasma mass spectroscopic detection. J Proteome Res 2010;9:3545–50. [280] Liu R, Xing Z, Lv Y, Zhang S, Zhang X. Sensitive sandwich immunoassay based on single particle mode inductively coupled plasma mass spectrometry detection. Talanta 2010;83:48–54. [281] Thompson DF, Eborall W, Dinsmore A, Smith CJ, Duckett CJ. Development and validation of a NANOGold™ immunoassay for the detection of vascular endothelial growth factor (VEGF) in human serum using inductively coupled plasma mass spectrometry. Rapid Commun Mass Spectrom 2010;24:927–32. [282] Liu J-M, Yan X-P. Ultrasensitive, selective and simultaneous detection of cytochrome c and insulin based on immunoassay and aptamer-based bioassay in combination with Au/Ag nanoparticle tagging and ICP-MS detection. J Anal At Spectrom 2011;26:1191–7. [283] Liu R, Liu X, Tang Y, Wu L, Hou X, Lv Y. Highly sensitive immunoassay based on immunogold–silver amplification and inductively coupled plasma mass spectrometric detection. Anal Chem 2011;83:2330–6. [284] He Q, Zhu Z, Jin L, Peng L, Guo W, Hu S. Detection of HIV-1 p24 antigen using streptavidin-biotin and gold nanoparticles based immunoassay by inductively coupled plasma mass spectrometry. J Anal At Spectrom 2014;29:1477–82. [285] Ko J, Lim HB. Multicore magnetic nanoparticles (MMNPs) doped with Cs and FITC for the determination of biomarker in serum using ICP-MS. Anal Chem 2014;86:4140–4. [286] Liu Z, Chen B, He M, Zhang X, Wang H, Hu B. Application of inductively coupled plasma mass spectrometry in the study of apoptosis: determination of caspase-3 using a gold nanoparticle tag. Analyst 2016;141: 926–33. [287] Li X, Chen B, He M, Xiao G, Hu B. Gold nanoparticle labeling with tyramide signal amplification for highly sensitive detection of alpha fetoprotein in human serum by ICP-MS. Talanta 2018;176:40–6. [288] Chen B, Peng H, Zheng F, Hu B, He M, Zhao W, et al. Immunoaffinity monolithic capillary microextraction coupled with ICP-MS for immunoassay with quantum dot labels. J Anal At Spectrom 2010;25:1674–81. [289] Tang Y, Jiao X, Liu R, Wu L, Wu L, Hou X, et al. Inductively coupled plasma mass spectrometry for determination of total urinary protein with CdTe quantum dots label. J Anal At Spectrom 2011;26:2493–9.

88

CHAPTER 2 NOVEL NANOMATERIALS FOR PROTEIN ANALYSIS

[290] Montoro Bustos AR, Garcia-Cortes M, Gonza´lez-Iglesias H, Ruiz Encinar J, Costa-Ferna´ndez JM, CocaPrados M, et al. Sensitive targeted multiple protein quantification based on elemental detection of Quantum Dots. Anal Chim Acta 2015;879:77–84. [291] Choi HW, Lee KH, Hur NH, Lim HB. Cerium oxide-deposited mesoporous silica nanoparticles for the determination of carcinoembryonic antigen in serum using inductively coupled plasma-mass spectrometry. Anal Chim Acta 2014;847:10–5. [292] Ko JA, Lim HB. Metal-doped inorganic nanoparticles for multiplex detection of biomarkers by a sandwichtype ICP-MS immunoassay. Anal Chim Acta 2016;938:1–6. [293] Cho HK, Lim HB. Determination of prostate-specific antigen (PSA) tagged with TiO2 nanoparticles using ICP-MS. J Anal At Spectrom 2013;28:468–72. [294] Chen B, Hu B, Jiang P, He M, Peng H, Zhang X. Nanoparticle labelling-based magnetic immunoassay on chip combined with electrothermal vaporization—inductively coupled plasma mass spectrometry for the determination of carcinoembryonic antigen in human serum. Analyst 2011;136:3934–42. [295] Liu Z, Yang B, Chen B, He M, Hu B. Upconversion nanoparticle as elemental tag for the determination of alpha-fetoprotein in human serum by inductively coupled plasma mass spectrometry. Analyst 2017;142: 197–205. [296] Chen Q, Yan G, Gao M, Zhang X. Ultrasensitive proteome profiling for 100 living cells by direct cell injection, online digestion and nano-LC-MS/MS analysis. Anal Chem 2015;87:6674–80.