Skip to main content

PERSPECTIVE article

Front. Anal. Sci., 22 June 2023
Sec. Omics
Volume 3 - 2023 | https://doi.org/10.3389/frans.2023.1186623

Toward the analysis of functional proteoforms using mass spectrometry-based stability proteomics

www.frontiersin.orgJi Kang www.frontiersin.orgMeena Seshadri www.frontiersin.orgKellye A. Cupp-Sutton* www.frontiersin.orgSi Wu*
  • Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, United States

Functional proteomics aims to elucidate biological functions, mechanisms, and pathways of proteins and proteoforms at the molecular level to examine complex cellular systems and disease states. A series of stability proteomics methods have been developed to examine protein functionality by measuring the resistance of a protein to chemical or thermal denaturation or proteolysis. These methods can be applied to measure the thermal stability of thousands of proteins in complex biological samples such as cell lysate, intact cells, tissues, and other biological fluids to measure proteome stability. Stability proteomics methods have been popularly applied to observe stability shifts upon ligand binding for drug target identification. More recently, these methods have been applied to characterize the effect of structural changes in proteins such as those caused by post-translational modifications (PTMs) and mutations, which can affect protein structures or interactions and diversify protein functions. Here, we discussed the current application of a suite of stability proteomics methods, including thermal proteome profiling (TPP), stability of proteomics from rates of oxidation (SPROX), and limited proteolysis (LiP) methods, to observe PTM-induced structural changes on protein stability. We also discuss future perspectives highlighting the integration of top-down mass spectrometry and stability proteomics methods to characterize intact proteoform stability and understand the function of variable protein modifications.

Introduction

Elucidating protein functionality is very important for understanding biological systems. The study of protein structure, function, and interactions has traditionally been done using classic structural biology methods, including protein nuclear magnetic resonance (NMR)(Yee et al, 2002), protein crystallography (Smyth and Martin, 2000), and cryogenic electron microscopy (cryo-EM)(Nickell et al, 2006). These methods are widely used, have high structural resolution, and can inform protein structural conformation as well as characterize interactions. Protein NMR has typically been applied for proteins from 10–30 kDa; however, some methods for protein NMR can be applied to very large proteins (>100 kDa) (Hu et al, 2021). Protein NMR can also assess protein structure and protein dynamics in solution and native-like environments. X-ray crystallography is the most common method for protein structural determination accounting for nearly 90% of structures in the PDB database (Benjin and Ling, 2020). More recent advances to Cryo-EM technology has allowed atomic resolution and made its use for protein structural determination more common (Yip et al, 2020).

However, despite these advantages, protein NMR and crystallography suffer from requirements for a relatively large amount of pure protein; time consuming protein expression and purification are often required. For X-ray crystallography, protein purification is followed by the need to grow protein crystals which can be challenging, particularly for disordered proteins and membrane proteins. Crystalline proteins may also have conformations that are not consistent with proteins in solution (Swaminathan et al, 2003; Acharya and Lloyd, 2005). Protein NMR often requires proteins to be expressed with heavy isotopes, uses solutions with high concentration (mM), and data analysis can be challenging and time-consuming (Venien-Bryan et al, 2016). Cryo-EM is currently unable to produce high resolution structures for proteins smaller than 30–50 kDa (D'Imprima and Kühlbrandt, 2021). Additionally, proteoforms with functional modifications such as post-translational modifications (PTMs) relevant to protein function generally cannot be expressed and purified as required for traditional structural biology approaches.

Overall, these structural biology methods for determination of protein function are robust and high resolution; however, drawbacks including low-throughput analysis of single proteins, requirement for large amounts of pure protein, and inability to characterize protein modifications has encouraged exploration into alternative methods for protein structural analysis. Hence, methods that provide structural and functional analysis of proteins using mass-spectrometry (MS)-based proteomics have been developed (Aebersold and Mann, 2003; Aebersold and Mann, 2016). While the resolution of these MS-based proteomics methods tends to be lower than the traditional structural biology methods discussed above, they benefit from high sensitivity, ability to analyze low purity samples or complex samples, and high throughput. Of particular interest is the ability of the MS-based methods to observe the structure and function of modified proteoforms (e.g., amino acid substitutions and post-translational modifications) that are often functionally relevant. MS-based proteomics methods capable of analyzing the structure, functions, and interactions of modified proteoforms include protein footprinting and stability proteomics methods.

A suite of MS-based protein footprinting methods, including Hydrogen-Deuterium Exchange mass spectrometry (HDX-MS) (Konermann et al, 2011), Fast Photochemical Oxidation of Protein (FPOP) (Johnson et al, 2019), Hydroxyl Radical Footprinting (HRF) (Xu et al, 2003), and Flash OXidation (FOX) (Sharp et al, 2021) have been developed to characterize functional proteins as well as their structures. Protein footprinting methods generally chemically label specific amino acid residues (or, in the case of HDX-MS, the amide backbone) that are solvent accessible in their native state to probe the protein composition, structure, and binding (Liu et al, 2020). Footprinting methods are generally low resolution when compared with the traditional structural biology methods discussed previously, typically achieving peptide-level resolution for HDX-MS analysis. Resolution for covalent labeling footprinting methods (excluding HDX-MS) can be limited by the existence of amino acid residues or functional groups appropriate for labeling. Initially, these protein footprinting methods were limited to pure protein and simple protein mixtures (Takamoto and Chance, 2006; Jones et al, 2011; Zhang et al, 2011; Sharp et al, 2021); however, more recent platforms such as those utilizing low-temperature LC separation for HDX-MS(Fang et al, 2021a) and more advanced data analysis techniques (Rinas and Jones, 2015) or in cell labeling (Kaur et al, 2020) for FPOP have enabled analysis of complex protein mixtures.

A series of MS-based proteomics methods, ‘stability proteomics’ has been developed to elucidate functional changes of proteins in complex mixtures (Kaur et al, 2018; Le Sueur et al, 2022). The tertiary structure of a protein is stabilized by various interactions within the proteins, including hydrophobic interactions, electrostatic interactions, hydrogen bonding, ionic bonding, and covalent bonding (disulfide bonds) (Gromiha, 2010; Pace et al, 2014). Stability proteomics generally falls into two categories: proteolysis-based methods and denaturation-based methods. Proteolysis-based methods, including Drug Affinity Responsive Target Stability (DARTS) (Lomenick et al, 2009) and Limited Proteolysis coupled with Mass Spectrometry (LiP-MS) (Feng et al, 2014), utilize the resistance of a protein to proteolysis to probe protein stability. Denaturation-based methods probe protein stability by studying the tendency of a protein to denature when exposed to a denaturing (chemical or thermal) gradient (Kaur et al, 2018). A few commonly applied denaturation-based methods include Stability of Proteins from Rates of Oxidation (SPROX) (West et al, 2008), Thermal Proteome Profiling (TPP) (Savitski et al, 2014), Chemical denaturation and Protein Precipitation (CPP) (Meng et al, 2018), Ion-based protein precipitation with Proteome-Integrated Solubility Alteration (i-PISA) (Beusch et al, 2022), and pulse proteolysis (Park and Marqusee, 2005; Chang et al, 2012).

These methods utilize the resistance of a protein to chemical or thermal denaturation or proteolysis to gauge protein stability. Protein stability can be altered by ligand binding (Celej et al, 2003; Majewski et al, 2019), protein-protein interactions (Layton and Hellinga, 2011), and protein modification (Le Sueur et al, 2022); as such, change in protein stability can be indicative of protein function (Teilum et al, 2011). The advantage of stability proteomics is the ability to analyze protein stability in complex protein mixtures (i.e., native or native-like environments) to observe proteome-level changes in protein stability. These methods have been used to observe the effect of ligand binding for drug target elucidation as well as to observe binding to non-target proteins, the effect of mutation and post-translational modification on proteome stability, and the effect of disease on proteome stability among other applications. Combinations of these techniques have also been evaluated to compare and contrast identified drug binding proteins between methods and compare proteome coverage (Cabrera et al, 2020; Xu et al, 2021). While these methods may offer limited (Strickland et al, 2013) or no resolution for the site of protein interactions, they provide a unique look at the functional proteome to observe unknown interactions and changes in protein stability.

As protein modifications due to alternative splicing, amino acid substitution, and post-translational modification are relevant to protein function by way of PTM crosstalk, protein-protein interactions, and protein activity (e.g., PTM switching (Ansong et al, 2013)), the application of these MS-based proteomics methods to modified functional proteoforms is of particular interest to researchers. In this review, we discuss the application of these stability proteomics methods (Supplementary Table S1), with a focus on TPP, to functionally modified and mutated proteoforms as well as the future perspectives regarding the application of stability proteomics methods to top-down proteomics.

Thermal proteome profiling

Among currently available stability proteomics methods, TPP which is based on the principles of the cellular thermal shift assay (CETSA) (Molina et al, 2013) is the most widely used to profile proteome-wide protein thermal stability of complex protein mixtures or intact cells (Savitski et al, 2014; Mateus et al, 2018; Mateus et al, 2020a). In a typical TPP experiment, aliquots of a sample (cells, cell lysate, tissue, etc.) are incubated at increasing temperatures, causing proteins to denature and precipitate at a specific temperature (melting point, Tm). Precipitates are removed and the soluble protein fractions are analyzed using bottom-up mass spectrometry with quantitative isobaric mass tag labeling and data dependent acquisition (Franken et al, 2015; Zhang et al, 2020) or label-free quantitation with data independent acquisition (George et al, 2023). Normalized protein abundance is plotted against the temperature, and the melting curve for each protein is fit using diverse methods/software packages that can determine melting point shifts and melting behavior similarities (Savitski et al, 2014; Leuenberger et al, 2017; Childs et al, 2019; Mateus et al, 2020a; Kurzawa et al, 2020; Fang et al, 2021b). Thermal proteome profiling has been used to study the melting behavior of proteins in situ under differing cellular conditions (Leuenberger et al, 2017; Mateus et al, 2020a).

TPP methods have been expanded and adapted for various purposes including drug target discovery and determination of binding strength. These methods include one-dimensional assays that utilize thermal gradients, concentration gradients, or isothermal dose-response (Franken et al, 2015; Lim et al, 2018). TPP methods have also been applied to many types of sample including intact cells, tissue, and whole blood (Perrin et al, 2020; Kalxdorf et al, 2021; Ruan et al, 2022; Zhao et al, 2022). Multidimensional methods that use both thermal and concentration gradients have also been developed to investigate thermal stability and binding affinity simultaneously (Becher et al, 2016; Mateus et al, 2018; Dziekan et al, 2020). Additionally, methods to probe the effect of protein expression, such as gene deletion in mutant strains (Mateus et al, 2020b), on protein expression and stability of the entire proteome have been developed. These are just a few of the interesting applications of TPP methods; others have been extensively reviewed in the literature (Mateus et al, 2017; Mateus et al, 2020a; de Souza and Picotti, 2020; Le Sueur et al, 2022; Mateus et al, 2022).

Recently, methods such as Hotspot Thermal Proteome Profiling (HTP) (Huang et al, 2019), Post-Translational Modification Thermal Proteome Profiling (PTM-TPP) (King et al, 2022), and Mutant Thermal Proteome Profiling (mTPP) (Peck Justice et al, 2020) were developed to assess changes in protein function due to structural changes such as variable PTMs and mutation (Figure 1). Similarly, System-wide Identification and prioritization of Enzyme Substrates by Thermal Analysis (SIESTA) was developed to identify and characterize enzyme substrates by observing the effect of enzyme catalyzed PTMs on substrate stability. (Saei et al, 2021). PTMs are covalent modifications of proteins that can alter protein function and increase the functional diversity of the proteome (Bagwan et al, 2021). Functionally relevant PTMs can affect protein function in a variety of ways; for example, modulation of protein-protein interactions (PPIs) (Seet et al, 2006; Duan and Walther, 2015), ATP production (Ardito et al, 2017), and function of cell surface and extracellular proteins in cancer and autoimmune disease (Reily et al, 2019). As such, high-throughput examination of PTM functionality has been a focus for advancement of the TPP methods discussed here.

FIGURE 1
www.frontiersin.org

FIGURE 1. Workflows for (A) phospho-TPP/hotspot thermal profiling (HTP), [schematic derived from Huang, J. X., et al. (2019)]; (B) post translational modification TPP (PTM-TPP) [schematic derived from King, D. T., et al. (2022)]; (C) mutant TPP (mTPP) [schematic derived from Peck Justice, S. A., et al. (2020)].

A subsect of TPP techniques, phospho-TPP (Potel et al, 2021; Smith et al, 2021) and HTP (Huang et al, 2019), have been developed to examine the effects of endogenous phosphorylation on thermal stability and protein functionality. Phosphorylation is of particular interest because it is an extremely common transient PTM that influences protein binding, stability, and dynamics (Johnson, 2009; Nishi et al, 2014). Combination of phosphopeptide enrichment strategies and TPP was first introduced by Azimi et al to determine the effect of protein phosphorylation on thermal stability of the phosphoproteome of SK-Mel 24 and SK-Mel 28 human carcinoma cells, which provided insights on protein interactions that could be targeted to counteract drug resistance of SK-Mel 28 (Azimi et al, 2018). This study demonstrated that these phospho-TPP methods enable determination of the effect of phosphorylation on thermal stability and protein function.

While analysis of the stability of HEK293T proteome by Huang et al did not report a significant difference in the Tm of the proteome and phosphoproteome, significant changes in the melting behavior for 25% of phosphorylated sites were reported (Huang et al, 2019). However, independent studies have shown that only 1.5%–3% of phosphosites significantly impact thermal protein stability (Potel et al, 2021; Lenz and Stühler, 2022). An improved HTP workflow involving high-pH reverse phase fractionation prior to phosphopeptide enrichment, label-fractionate-enrich (LFE)-HTP, has been reported for deeper phosphoproteome coverage (Stein et al, 2021).

TPP methods have also been applied to study the effect of PTMs such as O-GlcNAcylation on thermal stability (King et al, 2022). To analyze stability differences between modified proteoforms, two methods were used to observe the effect of O-GlcNAc on protein stability: enzymatic removal TPP (erPTM-TPP) and chemical modulators TPP (cmPTM-TPP). Though O-GlcNAc has often been reported to stabilize proteins (Drake et al, 2018), PTM-TPP methods found that O-GlcNAc tends to destabilize proteins due to interference in protein-protein interactions and its competition with protein phosphorylation (Issad et al, 2022).

Mutations of amino acids can disrupt protein function and cause disease by disrupting protein interactions (Zhong et al, 2009; Sahni et al, 2015), prevention of catalysis, allosteric regulation, disruption of a PTM, or, most commonly, changing the protein folding structure/stability (Wang and Moult, 2001; Bromberg and Rost, 2009). Additionally, site-directed mutagenesis is useful for interrupting standard gene activity and functional studies. As such, mutant TPP (mTPP) was developed as a high-throughput method to study the direct causes/pathways for genetic disorders and effects of site-directed mutagenesis and observe proteome-wide shifts in thermal stability resulting from amino acid substitution. mTPP was first introduced by Peck Justice et al (Peck Justice et al, 2020) to study temperature-sensitive (TS) mutants, specifically how changes in protein structure affected the thermal stability of the TS mutants compared to the unmodified protein and the PPI network as a whole. In a further application of mTPP by the Mosely group, (Peck Justice et al, 2021) applied affinity-purified protein complex as an isobaric trigger channel to determine the melting points of low abundance proteins.

Vieitez et al studied the functional relevance of phosphorylation on 474 phospho-deficient yeast strains (Viéitez et al, 2019). Screening their fitness in 102 different conditions showed changes in thermal stability in 8 phospho-mutants, with one exhibiting an increase and another a decrease in stability that was not statistically significant. (Banzhaf et al, 2020). studied the effect of an NlPI mutant on lipoproteins, finding that the deletion of NlPI altered protein abundance and thermal stability. While mTPP helps to understand the effect of mutations on protein stability, producing the TS mutants used in these studies often results in many mutations, making it difficult to determine which mutations cause which changes in protein stability. (Peck Justice et al, 2020).

In another interesting application of TPP, SIESTA utilizes the effect that PTMs have on protein thermal stability to identify known and unknown enzyme substrates (Saei et al, 2021). In SIESTA, lysate is incubated with an enzyme of interest and the enzyme cosubstrate to facilitate enzymatic post-translational modification of enzyme substrates. These modified substrates often demonstrate altered thermal stability resulting in an unbiased and high throughput method to identify enzyme substrates.

Overall, these methods depend on enrichment or overexpression of protein modifications to determine the effect of PTMs on protein stability. This is generally required as modified proteins tend to be lower in abundance than their modified proteoforms. However, very recently, (Kurzawa et al, 2023), introduced an alternative approach that applied a deep peptide identification method to TPP to observe the effect of modification on the thermal stability of coexisting functional proteoform groups. This technique did not require enrichment or overexpression of modified peptides and allowed more general analysis of changes in thermal stability caused by modifications such as PTMs, PPIs, etc.

Other stability proteomics methods for analysis of PTM induced stability shifts

While TPP is the most widely used stability proteomics technique, particularly for examining the effects of PTMs on protein stability, other stability proteomics methods, including SPROX and LiP, have been applied to this end. SPROX is unique in that it utilizes covalent labeling (e.g., methionine oxidation) to monitor protein unfolding (West et al, 2008). Proteins are unfolded using a chemical denaturant gradient and exposed to oxidizing conditions via the addition of hydrogen peroxide (H2O2) to selectively oxidize exposed methionine residues under each denaturant condition (Strickland et al, 2013). Oxidation is followed by a quenching agent (e.g., free methionine) to remove excess H2O2 and control labeling time. Proteins are subjected to digestion and methionine containing peptides are quantified using TMT labeling methods to determine the concentration at which proteins unfold and methionine residues become oxidized. SPROX also allows the examination of site-specific unfolding to determine the location of conformational changes or protein interactions.

LiP is another stability proteomics method that utilizes the resistance of a protein to proteolysis to determine protein stability (Feng et al, 2014). In LiP, proteins are digested with a nonspecific protease in the native state for a particular amount of time (Malinovska et al, 2023). Under these conditions, proteins with more accessible or unstructured regions tend to undergo more proteolysis than proteins with more stable structures. This reaction is quenched by heating and chemical denaturation, and the samples are further digested with a secondary protease such as trypsin. Tryptic and semitryptic/half-tryptic peptides are quantified using label-free MS-based methods to determine the extent to which a protein was digested in the native state.

SPROX and LiP have been applied to the analysis of the effect of phosphorylation on proteoform stability to study the significance of phosphorylation events in the MCF-7 breast cancer cell line, Figure 2 (Meng and Fitzgerald, 2018). MCF-7 cells were cultured in heavy and light SILAC media and the heavy lysate was treated with alkaline phosphatase to remove protein phosphorylation. These samples were subjected to LiP or SPROX workflows to determine changes in protein stability as a function of phosphorylation. Overall, SPROX and LiP found hundreds of proteins with changes in stability related to dephosphorylation. Additionally, these methods were able to identify changes in stability of proteins known to be differentially phosphorylated or stabilized in human breast cancer and proteins that are known to have function modulated by phosphorylation (Cappelletti et al, 2021) also applied LiP to the study of overall proteome stability in response to stress or nutrient adaptation. This study was also able to observe changes in protein stability due to phosphorylation in response to osmotic stress (Cappelletti et al, 2021).

FIGURE 2
www.frontiersin.org

FIGURE 2. Workflows for (A) post translational modification-limited proteolysis (PTM-LiP); (B) post translational modification stability of proteins from rates of oxidation (PTM-SPROX), [schematics derived from Meng, H, et al. (2018)].

While LiP and SPROX are the only stability proteomics methods, apart from TPP, that have been applied to the analysis of the effects of PTMs on protein stability, other methods such as pulse proteolysis could also potentially be applied for this purpose. Pulse proteolysis is similar to SPROX in that it probes protein stability using a chemical denaturation gradient (Park and Marqusee, 2005; Chang et al, 2012). Chemical denaturation is followed by a short proteolysis to digest denatured proteins.

Perspectives and future applications

Stability proteomics methods have seen an astonishing number of modifications and developments to make these techniques extremely versatile and applicable for a wide variety of purposes. The stability proteomics methods and applications reviewed here are unique in their goal of examining the effect of protein structural changes such as PTMs and mutation on protein stability or interaction. These methods have been applied with much success to determine the changes in stability caused by these modifications on the affected proteins directly, on protein interactions, and on the proteome generally. The bottom-up proteomics techniques currently applied to the stability proteomics methods reviewed here are robust, high-resolution, and have low limits of detection; however, the required proteolysis inhibits the examination of how structural changes affect intact proteoform stability (Catherman et al, 2014). As many PTMs of analogous and differing varieties can exist simultaneously on an intact proteoform to effect protein function and interactions, as is concisely demonstrated by the histone code (Jenuwein and Allis, 2001; Fornelli et al, 2018), protein digestion makes characterization of the intact, biologically active proteoform challenging. Furthermore, implementation of PTM specific enrichment strategies at the peptide level, as is done in many of the methods reviewed here, restricts the ability of these methods to observe how different numbers of PTM instances and combinations of different types of PTMs effect the stability of these proteoforms. As modified proteoforms can not only change the function of proteins but can also be the cause of or demonstrative of disease states, analysis of the total PTM profile of proteins is critical for biochemical analysis. As such, observation of the intact modified proteoforms directly can give a better understanding of how functional proteoforms perform and interact in vivo.

In contrast to bottom-up approaches, top-down proteomics approaches analyze intact proteins directly so that the structure of functional proteoforms can be examined (Gregorich and Ge, 2014). While there are several limitations to application of high-throughput top-down proteomics, a primary disadvantage is the lack of methods to assign functional annotations to intact proteoforms as recently reviewed by (Melby et al, 2021) This challenge stems from fundamental disadvantages in top-down proteomics including relatively low sensitivity hindering characterization of low abundance proteoforms, limitations in the size of proteoforms that can be analyzed using high resolution MS methods, and difficulty in applying quantitative proteomics methods to intact proteins. With these limitations, application of methods designed to probe protein functionality, such as stability proteomics, is very challenging using top-down methods. Fortunately, recent advancements in the area of top-down proteomics would allow application of some of these stability proteomics methods to examine how protein stability relates to unique protein proteoforms resulting in more detailed information regarding how protein modifications and combinations of modifications are involved in regulating protein function.

Intact proteoforms have many possible charge states resulting in wide charge state envelopes that decrease overall sensitivity. For high-throughput top-down proteomics, the effects of decreased sensitivity are made more prominent when proteins are coeluted from online separation methods which can result in ion suppression and difficulties in characterization, particularly of low abundance proteoforms (François et al, 2009; Pirok et al, 2019). Methods such as parallel ion parking (Chrisman et al, 2006) and ion/ion proton transfer (Stephenson and McLuckey, 1996) have been developed to narrow the charge envelope and increase sensitivity for MS analysis, and gas phase separation methods such as FAIMS have been implemented to decrease chemical noise and ion suppression (Gerbasi et al, 2021; Kaulich et al, 2022). Improving separation to decrease spectral complexity has also been implemented to increase depth of proteome analysis and improve intact proteoform characterization. In this vein, multidimensional separation is a technique that utilizes sequential, orthogonal separation techniques to improve separation resolution, proteome coverage, and characterization of proteins in a wide dynamic range. (Melby et al, 2021). However, the primary quantitative proteomics technique currently applied to top-down proteomics is label-free quantitation (Cupp-Sutton and Wu, 2020). As label-free quantitation requires an individual analysis for each experimental condition, it is not compatible with multidimensional separation. As such, application of other quantitative proteomics methods to top-down proteomics will improve quantitative accuracy and throughput.

Apart from label-free quantitation, the quantitative proteomics techniques currently applicable to top-down proteomics are metabolic labeling techniques and isobaric chemical tag labeling. (Cupp-Sutton and Wu, 2020). These techniques benefit from being compatible with multidimensional separation as well as having multiplex options to improve proteomic throughput. While metabolic labeling techniques have been applied for top-down proteomics, they must be implemented during cell growth to incorporate isotopic variants of amino acids into the protein primary structure (Ong et al, 2002). Therefore, metabolic labeling techniques are limited to samples that can be grown in lab and are not commonly used in stability proteomics methods. Isobaric chemical tag labeling, however, can be applied to any protein sample for flexible application. Until recently, isobaric chemical tag labeling techniques have not been widely applicable to high-throughput top-down proteomics due to issues with protein solubility and improper labeling under standard labeling conditions (Hung and Tholey, 2012). Of late, optimization of TMT labeling techniques including sample preparation and fragmentation have been developed for complex mixtures of intact proteins (Winkels et al, 2021; Yu et al, 2021; Guo et al, 2022a; Guo et al, 2022b; Guo et al, 2023). Application of these novel methods would allow for highly multiplexed TDP and application of multidimensional separations for improved quantitation and depth of proteome characterization.

Overall, developments in top-down proteomics including improved instrumental analysis, sample separation, and quantitation as discussed here have paved the way for application of top-down proteomics for more complex experimental designs such as stability proteomics methods. Stability proteomics techniques that do not require proteolysis (e.g., LiP, pulse proteolysis, DARTS) may be compatible with top-down proteomics, such as TPP, SPROX, CPP, PISA, and i-PISA. For example, TPP utilizes a thermal denaturation gradient to probe protein stability; therefore, the supernatant containing the soluble proteins can be directly analyzed using quantitative top-down proteomics approaches. MS-compatible buffers or online desalting techniques can be used to directly analyze TPP samples without additional sample preparation steps. Recently, the Wu lab has developed a label-free top-down TPP platform that has been applied to plot thermal profiles for intact proteoforms in complex samples such as Escherichia coli and human cell lysate (unpublished results). One possible challenge in analyzing these samples is the incomplete removal of precipitated proteins, which may cause column clogging, and improved methods for aggregate protein removal may be required. Applying top-down isobaric chemical tag labeling techniques can also improve the throughput of top-down TPP applications (Winkels et al, 2021; Yu et al, 2021; Guo et al, 2022a; Guo et al, 2022b; Guo et al, 2023).

Other methods such as SPROX, CPP, and i-PISA can also be coupled with top-down proteomics methods. However, these techniques use high concentrations of MS-incompatible buffers and reagents during sample preparation that can interfere with ion generation and lead to ion suppression during ESI and MS detection. For example, SPROX and CPP utilize high concentrations of chemical denaturants such as urea or guanidinium chloride to destabilize proteins, and i-PISA uses copper chloride for kosmotropic protein precipitation. Coupling existing methods (online or offline) for desalting intact protein samples (Cai et al, 2016) or protein precipitation and resolubilization [e.g., chloroform-methanol-water precipitation (Doucette et al, 2014)] would allow top-down proteomics analysis for these stability proteomics methods. These additional sample handling steps can lead to sample loss and increased error which decreases quantitative accuracy, particularly for label-free quantitation. Application of the intact protein isobaric chemical tag labeling protocol discussed above would necessitate buffer exchange prior to labeling regardless of MS compatibility of reagents used, and multiplexing limits run-to-run variability issues and reduces error to improve quantitation accuracy. Apart from these considerations, the application of top-down MS methods to stability proteomics methods largely overlap with bottom-up methods as demonstrated in Supplementary Table S1 including the resistance of proteins to certain types of denaturation and complexity in the denaturation behavior. To address some of these drawbacks, multiple proteomics methods have been applied to improve target identification and cross-verify identified targets (Cabrera et al, 2020; Bailey et al, 2023).

Overall, coupling top-down proteomics with the stability proteomics platforms reviewed here that focus on the characterization of the effect of protein modification on structural stability will open up the area of top-down proteomics for functional annotation of proteoforms, an area of research previously challenging to achieve.

Data availability statement

The original contributions presented in the study are included in the article/Supplementary Material, further inquiries can be directed to the corresponding author.

Author contributions

JK: Investigation, Visualization, Writing—Original draft preparation, KC-S: Conceptualization, Writing—Original draft preparation, SW: Conceptualization, Writing—Reviewing and Editing, Funding acquisition. All authors contributed to the article and approved the submitted version.

Funding

This work was partly supported by grants from OCAST HR23-169, NIH NIAID R01AI141625, and NIH NIH/NIAID2U19AI062629.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/frans.2023.1186623/full#supplementary-material

References

Acharya, K. R., and Lloyd, M. D., The advantages and limitations of protein crystal structures. Trends Pharmacol. Sci. (2005) 26(1):10–14. doi:10.1016/j.tips.2004.10.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Aebersold, R., and Mann, M. Mass spectrometry-based proteomics. Nature (2003) 422(6928):198–207. doi:10.1038/nature01511

PubMed Abstract | CrossRef Full Text | Google Scholar

Aebersold, R., and Mann, M. Mass-spectrometric exploration of proteome structure and function. Nature (2016) 537(7620):347–355. doi:10.1038/nature19949

PubMed Abstract | CrossRef Full Text | Google Scholar

Ansong, C., Wu, S., Meng, D., Liu, X., Brewer, H. M., Deatherage Kaiser, B. L., et al. Top-down proteomics reveals a unique protein S-thiolation switch in Salmonella Typhimurium in response to infection-like conditions. Proc. Natl. Acad. Sci. (2013) 110(25):10153–10158. doi:10.1073/pnas.1221210110

PubMed Abstract | CrossRef Full Text | Google Scholar

Ardito, F., Giuliani, M., Perrone, D., Troiano, G., and Lo Muzio, L. The crucial role of protein phosphorylation in cell signaling and its use as targeted therapy (Review). Int. J. Mol. Med. (2017) 40(2):271–280. doi:10.3892/ijmm.2017.3036

PubMed Abstract | CrossRef Full Text | Google Scholar

Azimi, A., Caramuta, S., Seashore-Ludlow, B., Boström, J., Robinson, J. L., Edfors, F., et al. Targeting CDK2 overcomes melanoma resistance against BRAF and Hsp90 inhibitors. Mol. Syst. Biol. (2018) 14(3):e7858. doi:10.15252/msb.20177858

PubMed Abstract | CrossRef Full Text | Google Scholar

Bagwan, N., El Ali, H. H., and Lundby, A. Proteome-wide profiling and mapping of post translational modifications in human hearts. Sci. Rep. (2021) 11(1):2184. doi:10.1038/s41598-021-81986-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Bailey, M. A., Tang, Y., Park, H.-J., and Fitzgerald, M. C. Comparative analysis of protein folding stability-based profiling methods for characterization of biological phenotypes. J. Am. Soc. Mass Spectrom. (2023) 34(3):383–393. doi:10.1021/jasms.2c00248

PubMed Abstract | CrossRef Full Text | Google Scholar

Banzhaf, M., Yau, H. C., Verheul, J., Lodge, A., Kritikos, G., Mateus, A., et al. Outer membrane lipoprotein NlpI scaffolds peptidoglycan hydrolases within multi-enzyme complexes in Escherichia coli. EMBO J. (2020) 39(5):e102246. doi:10.15252/embj.2019102246

PubMed Abstract | CrossRef Full Text | Google Scholar

Becher, I., Werner, T., Doce, C., Zaal, E. A., Tögel, I., Khan, C. A., et al. Thermal profiling reveals phenylalanine hydroxylase as an off-target of panobinostat. Nat. Chem. Biol. (2016) 12(11):908–910. doi:10.1038/nchembio.2185

PubMed Abstract | CrossRef Full Text | Google Scholar

Benjin, X., and Ling, L. Developments, applications, and prospects of cryo-electron microscopy. Protein Sci. (2020) 29(4):872–882. doi:10.1002/pro.3805

PubMed Abstract | CrossRef Full Text | Google Scholar

Beusch, C. M., Sabatier, P., and Zubarev, R. A. Ion-based proteome-integrated solubility alteration assays for systemwide profiling of protein–molecule interactions. Anal. Chem. (2022) 94(19):7066–7074. doi:10.1021/acs.analchem.2c00391

PubMed Abstract | CrossRef Full Text | Google Scholar

Bromberg, Y., and Rost, B. Correlating protein function and stability through the analysis of single amino acid substitutions. BMC Bioinforma. (2009) 10(8):S8. doi:10.1186/1471-2105-10-S8-S8

CrossRef Full Text | Google Scholar

Cabrera, A., Wiebelhaus, N., Quan, B., Ma, R., Meng, H., and Fitzgerald, M. C. Comparative analysis of mass-spectrometry-based proteomic methods for protein target discovery using a one-pot approach. J. Am. Soc. Mass Spectrom. (2020) 31(2):217–226. doi:10.1021/jasms.9b00041

PubMed Abstract | CrossRef Full Text | Google Scholar

Cai, W., Tucholski, T. M., Gregorich, Z. R., and Ge, Y. Top-down proteomics: Technology advancements and applications to heart diseases. Expert Rev. Proteomics (2016) 13(8):717–730. doi:10.1080/14789450.2016.1209414

PubMed Abstract | CrossRef Full Text | Google Scholar

Cappelletti, V., Hauser, T., Piazza, I., Pepelnjak, M., Malinovska, L., Fuhrer, T., et al. Dynamic 3D proteomes reveal protein functional alterations at high resolution in situ. Cell (2021) 184(2):545–559.e22. doi:10.1016/j.cell.2020.12.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Catherman, A. D., Skinner, O. S., and Kelleher, N. L. Top down proteomics: Facts and perspectives. Biochem. Biophysical Res. Commun. (2014) 445(4):683–693. doi:10.1016/j.bbrc.2014.02.041

CrossRef Full Text | Google Scholar

Celej, M. S., Montich, G. G., and Fidelio, G. D. Protein stability induced by ligand binding correlates with changes in protein flexibility. Protein Sci. (2003) 12(7):1496–1506. doi:10.1110/ps.0240003

PubMed Abstract | CrossRef Full Text | Google Scholar

Chang, Y., Schlebach, J. P., VerHeul, R. A., and Park, C. Simplified proteomics approach to discover protein–ligand interactions. Protein Sci. (2012) 21(9):1280–1287. doi:10.1002/pro.2112

PubMed Abstract | CrossRef Full Text | Google Scholar

Childs, D., Bach, K., Franken, H., Anders, S., Kurzawa, N., Bantscheff, M., et al. Nonparametric analysis of thermal proteome profiles reveals novel drug-binding proteins. Mol. Cell. Proteomics (2019) 18(12):2506–2515. doi:10.1074/mcp.TIR119.001481

PubMed Abstract | CrossRef Full Text | Google Scholar

Chrisman, P. A., Pitteri, S. J., and McLuckey, S. A. Parallel ion parking of protein mixtures. Anal. Chem. (2006) 78(1):310–316. doi:10.1021/ac0515778

PubMed Abstract | CrossRef Full Text | Google Scholar

Cupp-Sutton, K. A., and Wu, S. High-throughput quantitative top-down proteomics. Mol. Omics (2020) 16:91–99. doi:10.1039/c9mo00154a

PubMed Abstract | CrossRef Full Text | Google Scholar

D'Imprima, E., and Kühlbrandt, W. Current limitations to high-resolution structure determination by single-particle cryoEM. Q. Rev. Biophysics (2021) 54:e4. doi:10.1017/S0033583521000020

CrossRef Full Text | Google Scholar

de Souza, N., and Picotti, P. Mass spectrometry analysis of the structural proteome. Curr. Opin. Struct. Biol. (2020) 60:57–65. doi:10.1016/j.sbi.2019.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Doucette, A. A., Vieira, D. B., Orton, D. J., and Wall, M. J. Resolubilization of precipitated intact membrane proteins with cold formic acid for analysis by mass spectrometry. J. Proteome Res. (2014) 13(12):6001–6012. doi:10.1021/pr500864a

PubMed Abstract | CrossRef Full Text | Google Scholar

Drake, W. R., Hou, C.-W., Zachara, N. E., and Grimes, C. L. New use for CETSA: Monitoring innate immune receptor stability via post-translational modification by OGT. J. Bioenergetics Biomembr. (2018) 50(3):231–240. doi:10.1007/s10863-018-9754-z

CrossRef Full Text | Google Scholar

Duan, G., and Walther, D. The roles of post-translational modifications in the context of protein interaction networks. PLOS Comput. Biol. (2015) 11(2):e1004049. doi:10.1371/journal.pcbi.1004049

PubMed Abstract | CrossRef Full Text | Google Scholar

Dziekan, J. M., Wirjanata, G., Dai, L., Go, K. D., Yu, H., Lim, Y. T., et al. Cellular thermal shift assay for the identification of drug–target interactions in the Plasmodium falciparum proteome. Nat. Protoc. (2020) 15(6):1881–1921. doi:10.1038/s41596-020-0310-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, M., Wang, Z., Cupp-Sutton, K. A., Welborn, T., Smith, K., and Wu, S. High-throughput hydrogen deuterium exchange mass spectrometry (HDX-MS) coupled with subzero-temperature ultrahigh pressure liquid chromatography (UPLC) separation for complex sample analysis. Anal. Chim. Acta (2021) 1143:65–72. doi:10.1016/j.aca.2020.11.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, S., Kirk, P. D. W., Bantscheff, M., Lilley, K. S., and Crook, O. M. A Bayesian semi-parametric model for thermal proteome profiling. Commun. Biol. (2021) 4(1):810. doi:10.1038/s42003-021-02306-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Feng, Y., De Franceschi, G., Kahraman, A., Soste, M., Melnik, A., Boersema, P. J., et al. Global analysis of protein structural changes in complex proteomes. Nat. Biotechnol. (2014) 32(10):1036–1044. doi:10.1038/nbt.2999

PubMed Abstract | CrossRef Full Text | Google Scholar

Fornelli, L., Toby, T. K., Schachner, L. F., Doubleday, P. F., Srzentić, K., DeHart, C. J., et al. Top-down proteomics: Where we are, where we are going? J. Proteomics (2018) 175:3–4. doi:10.1016/j.jprot.2017.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

François, I., Sandra, K., and Sandra, P. Comprehensive liquid chromatography: Fundamental aspects and practical considerations—a review. Anal. Chim. Acta (2009) 641(1):14–31. doi:10.1016/j.aca.2009.03.041

PubMed Abstract | CrossRef Full Text | Google Scholar

Franken, H., Mathieson, T., Childs, D., Sweetman, G. M. A., Werner, T., Toegel, I., et al. Thermal proteome profiling for unbiased identification of direct and indirect drug targets using multiplexed quantitative mass spectrometry. Nat. Protoc. (2015) 10(10):1567–1593. doi:10.1038/nprot.2015.101

PubMed Abstract | CrossRef Full Text | Google Scholar

George, A. L., Sidgwick, F. R., Watt, J. E., Martin, M. P., Trost, M., Marín-Rubio, J. L., et al. A comparison of quantitative mass spectrometric methods for drug target identification by thermal proteome profiling. bioRxiv (2023) 2023. doi:10.1101/2023.02.15.528618

CrossRef Full Text | Google Scholar

Gerbasi, V. R., Melani, R. D., Abbatiello, S. E., Belford, M. W., Huguet, R., McGee, J. P., et al. Deeper protein identification using field asymmetric ion mobility spectrometry in top-down proteomics. Anal. Chem. (2021) 93(16):6323–6328. doi:10.1021/acs.analchem.1c00402

PubMed Abstract | CrossRef Full Text | Google Scholar

Gregorich, Z. R., and Ge, Y. Top-down proteomics in health and disease: Challenges and opportunities. PROTEOMICS (2014) 14(10):1195–1210. doi:10.1002/pmic.201300432

PubMed Abstract | CrossRef Full Text | Google Scholar

Gromiha, M. M. Protein stability. Amsterdam, Netherlands; Elsevier (2010).

Google Scholar

Guo, Y., Chowdhury, T., Seshadri, M., Cupp-Sutton, K. A., Wang, Q., Yu, D., et al. Optimization of higher-energy collisional dissociation fragmentation energy for intact protein-level tandem mass tag labeling. J. Proteome Res. (2023) 22:1406–1418. doi:10.1021/acs.jproteome.2c00549

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, Y., Yu, D., Cupp-Sutton, K. A., Liu, X., and Wu, S. A benchmarking protocol for intact protein-level Tandem Mass Tag (TMT) labeling for quantitative top-down proteomics. MethodsX (2022) 9:101873. doi:10.1016/j.mex.2022.101873

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, Y., Yu, D., Cupp-Sutton, K. A., Liu, X., and Wu, S. Optimization of protein-level tandem mass tag (TMT) labeling conditions in complex samples with top-down proteomics. Anal. Chim. Acta (2022) 1221:340037. doi:10.1016/j.aca.2022.340037

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, Y., Cheng, K., He, L., Zhang, X., Jiang, B., Jiang, L., et al. NMR-based methods for protein analysis. Anal. Chem. (2021) 93(4):1866–1879. doi:10.1021/acs.analchem.0c03830

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, J. X., Lee, G., Cavanaugh, K. E., Chang, J. W., Gardel, M. L., and Moellering, R. E. High throughput discovery of functional protein modifications by Hotspot Thermal Profiling. Nat. Methods (2019) 16(9):894–901. doi:10.1038/s41592-019-0499-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Hung, C.-W., and Tholey, A. Tandem mass tag protein labeling for top-down identification and quantification. Anal. Chem. Wash. D.C. U. S.) (2012) 84(1):161–170. doi:10.1021/ac202243r

CrossRef Full Text | Google Scholar

Issad, T., Al-Mukh, H., Bouaboud, A., and Pagesy, P. Protein O-GlcNAcylation and the regulation of energy homeostasis: Lessons from knock-out mouse models. J. Biomed. Sci. (2022) 29(1):64. doi:10.1186/s12929-022-00851-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Jenuwein, T., and Allis, C. D. Translating the histone code. Science (2001) 293(5532):1074–1080. doi:10.1126/science.1063127

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnson, D. T., Di Stefano, L. H., and Jones, L. M. Fast photochemical oxidation of proteins (FPOP): A powerful mass spectrometry–based structural proteomics tool. J. Biol. Chem. (2019) 294(32):11969–11979. doi:10.1074/jbc.REV119.006218

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnson, Louise N. The regulation of protein phosphorylation. Biochem. Soc. Trans. (2009) 37(4):627–641. doi:10.1042/BST0370627

PubMed Abstract | CrossRef Full Text | Google Scholar

Jones, L. M., Sperry, J. B., Carroll, J. A., and Gross, M. L. Fast photochemical oxidation of proteins for epitope mapping. Anal. Chem. (2011) 83(20):7657–7661. doi:10.1021/ac2007366

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalxdorf, M., Günthner, I., Becher, I., Kurzawa, N., Knecht, S., Savitski, M. M., et al. Cell surface thermal proteome profiling tracks perturbations and drug targets on the plasma membrane. Nat. Methods (2021) 18(1):84–91. doi:10.1038/s41592-020-01022-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaulich, P. T., Cassidy, L., Winkels, K., and Tholey, A. Improved identification of proteoforms in top-down proteomics using FAIMS with internal CV stepping. Anal. Chem. (2022) 94(8):3600–3607. doi:10.1021/acs.analchem.1c05123

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaur, U., Johnson, D. T., and Jones, L. M. Validation of the applicability of in-cell Fast photochemical oxidation of proteins across multiple eukaryotic cell lines. J. Am. Soc. Mass Spectrom. (2020) 31(7):1372–1379. doi:10.1021/jasms.0c00014

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaur, U., Meng, H., Lui, F., Ma, R., Ogburn, R. N., Johnson, J. H. R., et al. Proteome-wide structural biology: An emerging field for the structural analysis of proteins on the proteomic scale. J. Proteome Res. (2018) 17(11):3614–3627. doi:10.1021/acs.jproteome.8b00341

PubMed Abstract | CrossRef Full Text | Google Scholar

King, D. T., Serrano-Negrón, J. E., Zhu, Y., Moore, C. L., Shoulders, M. D., Foster, L. J., et al. Thermal proteome profiling reveals the O-GlcNAc-Dependent meltome. J. Am. Chem. Soc. (2022) 144(9):3833–3842. doi:10.1021/jacs.1c10621

PubMed Abstract | CrossRef Full Text | Google Scholar

Konermann, L., Pan, J., and Liu, Y.-H. Hydrogen exchange mass spectrometry for studying protein structure and dynamics. Chem. Soc. Rev. (2011) 40(3):1224–1234. doi:10.1039/c0cs00113a

PubMed Abstract | CrossRef Full Text | Google Scholar

Kurzawa, N., Becher, I., Sridharan, S., Franken, H., Mateus, A., Anders, S., et al. A computational method for detection of ligand-binding proteins from dose range thermal proteome profiles. Nat. Commun. (2020) 11(1):5783. doi:10.1038/s41467-020-19529-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Kurzawa, N., Leo, I. R., Stahl, M., Kunold, E., Becher, I., Audrey, A., et al. Deep thermal profiling for detection of functional proteoform groups. Nat. Chem. Biol. (2023). doi:10.1038/s41589-023-01284-8

CrossRef Full Text | Google Scholar

Layton, C. J., and Hellinga, H. W. Quantitation of protein–protein interactions by thermal stability shift analysis. Protein Sci. (2011) 20(8):1439–1450. doi:10.1002/pro.674

PubMed Abstract | CrossRef Full Text | Google Scholar

Le Sueur, C., Hammarén, H. M., Sridharan, S., and Savitski, M. M. Thermal proteome profiling: Insights into protein modifications, associations, and functions. Curr. Opin. Chem. Biol. (2022) 71:102225. doi:10.1016/j.cbpa.2022.102225

PubMed Abstract | CrossRef Full Text | Google Scholar

Lenz, T., and Stühler, K. Small molecule arranged thermal proximity coaggregation (smarTPCA)—a novel approach to characterize protein–protein interactions in living cells by similar isothermal dose–responses. Int. J. Mol. Sci. (2022) 23(10):5605. doi:10.3390/ijms23105605

PubMed Abstract | CrossRef Full Text | Google Scholar

Leuenberger, P., Ganscha, S., Kahraman, A., Cappelletti, V., Boersema, P. J., von Mering, C., et al. Cell-wide analysis of protein thermal unfolding reveals determinants of thermostability. Science (2017) 355(6327):eaai7825. doi:10.1126/science.aai7825

PubMed Abstract | CrossRef Full Text | Google Scholar

Lim, Y. T., Prabhu, N., Dai, L., Go, K. D., Chen, D., Sreekumar, L., et al. An efficient proteome-wide strategy for discovery and characterization of cellular nucleotide-protein interactions. PLOS ONE (2018) 13(12):e0208273. doi:10.1371/journal.pone.0208273

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X. R., Zhang, M. M., and Gross, M. L. Mass spectrometry-based protein footprinting for higher-order structure analysis: Fundamentals and applications. Chem. Rev. (2020) 120(10):4355–4454. doi:10.1021/acs.chemrev.9b00815

PubMed Abstract | CrossRef Full Text | Google Scholar

Lomenick, B., Hao, R., Jonai, N., Chin, R. M., Aghajan, M., Warburton, S., et al. Target identification using drug affinity responsive target stability (DARTS). Proc. Natl. Acad. Sci. (2009) 106(51):21984–21989. doi:10.1073/pnas.0910040106

PubMed Abstract | CrossRef Full Text | Google Scholar

Majewski, M., Ruiz-Carmona, S., and Barril, X. An investigation of structural stability in protein-ligand complexes reveals the balance between order and disorder. Commun. Chem. (2019) 2(1):110. doi:10.1038/s42004-019-0205-5

CrossRef Full Text | Google Scholar

Malinovska, L., Cappelletti, V., Kohler, D., Piazza, I., Tsai, T.-H., Pepelnjak, M., et al. Proteome-wide structural changes measured with limited proteolysis-mass spectrometry: An advanced protocol for high-throughput applications. Nat. Protoc. (2023) 18(3):659–682. doi:10.1038/s41596-022-00771-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Mateus, A., Bobonis, J., Kurzawa, N., Hevler, J., Typas, A., Savitski, M. M., et al. Thermal proteome profiling in bacteria: Probing protein state in vivo. Mol. Syst. Biol. (2018) 14(7):e8242. doi:10.15252/msb.20188242

PubMed Abstract | CrossRef Full Text | Google Scholar

Mateus, A., Hevler, J., Bobonis, J., Kurzawa, N., Shah, M., Mitosch, K., et al. The functional proteome landscape of Escherichia coli. Nature (2020) 588(7838):473–478. doi:10.1038/s41586-020-3002-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Mateus, A., Kurzawa, N., Becher, I., Sridharan, S., Helm, D., Stein, F., et al. Thermal proteome profiling for interrogating protein interactions. Mol. Syst. Biol. (2020) 16(3):e9232. doi:10.15252/msb.20199232

PubMed Abstract | CrossRef Full Text | Google Scholar

Mateus, A., Kurzawa, N., Perrin, J., Bergamini, G., and Savitski, M. M. Drug target identification in tissues by thermal proteome profiling. Annu. Rev. Pharmacol. Toxicol. (2022) 62(1):465–482. doi:10.1146/annurev-pharmtox-052120-013205

PubMed Abstract | CrossRef Full Text | Google Scholar

Mateus, A., Määttä, T. A., and Savitski, M. M. Thermal proteome profiling: Unbiased assessment of protein state through heat-induced stability changes. Proteome Sci. (2017) 15(1):13. doi:10.1186/s12953-017-0122-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Melby, J. A., Roberts, D. S., Larson, E. J., Brown, K. A., Bayne, E. F., Jin, S., et al. Novel strategies to address the challenges in top-down proteomics. J. Am. Soc. Mass Spectrom. (2021) 32(6):1278–1294. doi:10.1021/jasms.1c00099

PubMed Abstract | CrossRef Full Text | Google Scholar

Meng, H., and Fitzgerald, M. C. Proteome-wide characterization of phosphorylation-induced conformational changes in breast cancer. J. Proteome Res. (2018) 17(3):1129–1137. doi:10.1021/acs.jproteome.7b00795

PubMed Abstract | CrossRef Full Text | Google Scholar

Meng, H., Ma, R., and Fitzgerald, M. C. Chemical denaturation and protein precipitation approach for discovery and quantitation of protein-drug interactions. Anal. Chem. Wash. D.C. U. S.) (2018) 90(15):9249–9255. doi:10.1021/acs.analchem.8b01772

CrossRef Full Text | Google Scholar

Molina, D. M., Jafari, R., Ignatushchenko, M., Seki, T., Larsson, E. A., Dan, C., et al. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science (2013) 341(6141):84–87. doi:10.1126/science.1233606

PubMed Abstract | CrossRef Full Text | Google Scholar

Nickell, S., Kofler, C., Leis, A. P., and Baumeister, W. A visual approach to proteomics. Nat. Rev. Mol. Cell Biol. (2006) 7(3):225–230. doi:10.1038/nrm1861

PubMed Abstract | CrossRef Full Text | Google Scholar

Nishi, H., Shaytan, A., and Panchenko, A. R. Physicochemical mechanisms of protein regulation by phosphorylation. Front. Genet. (2014) 5:270. doi:10.3389/fgene.2014.00270

PubMed Abstract | CrossRef Full Text | Google Scholar

Ong, S.-E., Blagoev, B., Kratchmarova, I., Kristensen, D. B., Steen, H., Pandey, A., et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteomics (2002) 1(5):376–386. doi:10.1074/mcp.m200025-mcp200

PubMed Abstract | CrossRef Full Text | Google Scholar

Pace, C. N., Fu, H., Lee Fryar, K., Landua, J., Trevino, S. R., Schell, D., et al. Contribution of hydrogen bonds to protein stability. Protein Sci. (2014) 23(5):652–661. doi:10.1002/pro.2449

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, C., and Marqusee, S. Pulse proteolysis: A simple method for quantitative determination of protein stability and ligand binding. Nat. Methods (2005) 2(3):207–212. doi:10.1038/nmeth740

PubMed Abstract | CrossRef Full Text | Google Scholar

Peck Justice, S. A., Barron, M. P., Qi, G. D., Wijeratne, H. R. S., Victorino, J. F., Simpson, E. R., et al. Mutant thermal proteome profiling for characterization of missense protein variants and their associated phenotypes within the proteome. J. Biol. Chem. (2020) 295(48):16219–16238. doi:10.1074/jbc.RA120.014576

PubMed Abstract | CrossRef Full Text | Google Scholar

Peck Justice, S. A., McCracken, N. A., Victorino, J. F., Qi, G. D., Wijeratne, A. B., and Mosley, A. L. Boosting detection of low-abundance proteins in thermal proteome profiling experiments by addition of an isobaric trigger channel to TMT multiplexes. Anal. Chem. (2021) 93(18):7000–7010. doi:10.1021/acs.analchem.1c00012

PubMed Abstract | CrossRef Full Text | Google Scholar

Perrin, J., Werner, T., Kurzawa, N., Rutkowska, A., Childs, D. D., Kalxdorf, M., et al. Identifying drug targets in tissues and whole blood with thermal-shift profiling. Nat. Biotechnol. (2020) 38(3):303–308. doi:10.1038/s41587-019-0388-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Pirok, B. W. J., Stoll, D. R., and Schoenmakers, P. J. Recent developments in two-dimensional liquid chromatography: Fundamental improvements for practical applications. Anal. Chem. (2019) 91(1):240–263. doi:10.1021/acs.analchem.8b04841

PubMed Abstract | CrossRef Full Text | Google Scholar

Potel, C. M., Kurzawa, N., Becher, I., Typas, A., Mateus, A., and Savitski, M. M. Impact of phosphorylation on thermal stability of proteins. Nat. Methods (2021) 18(7):757–759. doi:10.1038/s41592-021-01177-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Reily, C., Stewart, T. J., Renfrow, M. B., and Novak, J. Glycosylation in health and disease. Nat. Rev. Nephrol. (2019) 15(6):346–366. doi:10.1038/s41581-019-0129-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Rinas, A., and Jones, L. M. Fast photochemical oxidation of proteins coupled to multidimensional protein identification technology (MudPIT): Expanding footprinting strategies to complex systems. J. Am. Soc. Mass Spectrom. (2015) 26(4):540–546. doi:10.1007/s13361-014-1017-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruan, C., Ning, W., Liu, Z., Zhang, X., Fang, Z., Li, Y., et al. Precipitate-supported thermal proteome profiling coupled with deep learning for comprehensive screening of drug target proteins. ACS Chem. Biol. (2022) 17(1):252–262. doi:10.1021/acschembio.1c00936

PubMed Abstract | CrossRef Full Text | Google Scholar

Saei, A. A., Beusch, C. M., Sabatier, P., Wells, J. A., Gharibi, H., Meng, Z., et al. System-wide identification and prioritization of enzyme substrates by thermal analysis. Nat. Commun. (2021) 12(1):1296. doi:10.1038/s41467-021-21540-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Sahni, N., Yi, S., Taipale, M., Bass, F., Juan, I., Coulombe-Huntington, J., et al. Widespread macromolecular interaction perturbations in human genetic disorders. Cell (2015) 161(3):647–660. doi:10.1016/j.cell.2015.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Savitski, M. M., Reinhard, F. B. M., Franken, H., Werner, T., Savitski, M. F., Eberhard, D., et al. Tracking cancer drugs in living cells by thermal profiling of the proteome. Science (2014) 346(6205):1255784. doi:10.1126/science.1255784

PubMed Abstract | CrossRef Full Text | Google Scholar

Seet, B. T., Dikic, I., Zhou, M.-M., and Pawson, T. Reading protein modifications with interaction domains. Nat. Rev. Mol. Cell Biol. (2006) 7(7):473–483. doi:10.1038/nrm1960

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharp, J. S., Chea, E. E., Misra, S. K., Orlando, R., Popov, M., Egan, R. W., et al. Flash oxidation (FOX) system: A novel laser-free Fast photochemical oxidation protein footprinting platform. J. Am. Soc. Mass Spectrom. (2021) 32(7):1601–1609. doi:10.1021/jasms.0c00471

PubMed Abstract | CrossRef Full Text | Google Scholar

Smith, I. R., Hess, K. N., Bakhtina, A. A., Valente, A. S., Rodríguez-Mias, R. A., and Villén, J. Identification of phosphosites that alter protein thermal stability. Nat. Methods (2021) 18(7):760–762. doi:10.1038/s41592-021-01178-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Smyth, M. S., and Martin, J. H. J. x Ray crystallography. Mol. Pathol. (2000) 53(1):8–14. doi:10.1136/mp.53.1.8

PubMed Abstract | CrossRef Full Text | Google Scholar

Stein, B. D., Huang, J. X., Wu, D., Cantley, L. C., and Moellering, R. E. Diverse hotspot thermal profiling methods detect phosphorylation-dependent changes in protein stability. bioRxiv (2021) 2021. doi:10.1101/2021.05.01.441686

CrossRef Full Text | Google Scholar

Stephenson, J. L., and McLuckey, S. A. Ion/ion proton transfer reactions for protein mixture analysis. Anal. Chem. (1996) 68(22):4026–4032. doi:10.1021/ac9605657

PubMed Abstract | CrossRef Full Text | Google Scholar

Strickland, E. C., Geer, M. A., Tran, D. T., Adhikari, J., West, G. M., DeArmond, P. D., et al. Thermodynamic analysis of protein-ligand binding interactions in complex biological mixtures using the stability of proteins from rates of oxidation. Nat. Protoc. (2013) 8(1):148–161. doi:10.1038/nprot.2012.146

PubMed Abstract | CrossRef Full Text | Google Scholar

Swaminathan, G. J., Holloway, D. E., Colvin, R. A., Campanella, G. K., Papageorgiou, A. C., Luster, A. D., et al. Crystal structures of oligomeric forms of the IP-10/CXCL10 chemokine. Structure (2003) 11(5):521–532. doi:10.1016/s0969-2126(03)00070-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Takamoto, K., and Chance, M. R. Radiolytic protein footprinting with mass spectrometry to probe the structure of macromolecular complexes. Annu. Rev. Biophysics Biomol. Struct. (2006) 35(1):251–276. doi:10.1146/annurev.biophys.35.040405.102050

CrossRef Full Text | Google Scholar

Teilum, K., Olsen, J. G., and Kragelund, B. B. Protein stability, flexibility and function. Biochimica Biophysica Acta (BBA) - Proteins Proteomics (2011) 1814(8):969–976. doi:10.1016/j.bbapap.2010.11.005

CrossRef Full Text | Google Scholar

Venien-Bryan, C., Li, Z., Vuillard, L., and Boutin, J. A. (2016). Cryo-electron microscopy and X-ray crystallography: Complementary approaches to structural biology and drug discovery, Acta Crystallogr. Sect. F., 73 (4), MedSci. (Paris), 32, 758-767] 174–183. doi:10.1107/S2053230X17003740

CrossRef Full Text | Google Scholar

Viéitez, C., Busby, B. P., Ochoa, D., Mateus, A., Galardini, M., Jawed, A., et al. Towards a systematic map of the functional role of protein phosphorylation. bioRxiv (2019) 872770.

Google Scholar

Wang, Z., and Moult, J. SNPs, protein structure, and disease. Hum. Mutat. (2001) 17(4):263–270. doi:10.1002/humu.22

PubMed Abstract | CrossRef Full Text | Google Scholar

West, G. M., Tang, L., and Fitzgerald, M. C. Thermodynamic analysis of protein stability and ligand binding using a chemical modification- and mass spectrometry-based strategy. Anal. Chem. Wash. D.C. U. S.) (2008) 80(11):4175–4185. doi:10.1021/ac702610a

CrossRef Full Text | Google Scholar

Winkels, K., Koudelka, T., and Tholey, A. Quantitative top-down proteomics by isobaric labeling with thiol-directed tandem mass tags. J. Proteome Res. (2021) 20(9):4495–4506. doi:10.1021/acs.jproteome.1c00460

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, G., Takamoto, K., and Chance, M. R. Radiolytic modification of basic amino acid residues in peptides: Probes for examining Protein−Protein interactions. Anal. Chem. (2003) 75(24):6995–7007. doi:10.1021/ac035104h

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, Y., West, G. M., Abdelmessih, M., Troutman, M. D., and Everley, R. A. A comparison of two stability proteomics methods for drug target identification in OnePot 2D format. ACS Chem. Biol. (2021) 16(8):1445–1455. doi:10.1021/acschembio.1c00317

PubMed Abstract | CrossRef Full Text | Google Scholar

Yee, A., Chang, X., Pineda-Lucena, A., Wu, B., Semesi, A., Le, B., et al. An NMR approach to structural proteomics. Proc. Natl. Acad. Sci. (2002) 99(4):1825–1830. doi:10.1073/pnas.042684599

PubMed Abstract | CrossRef Full Text | Google Scholar

Yip, K. M., Fischer, N., Paknia, E., Chari, A., and Stark, H. Atomic-resolution protein structure determination by cryo-EM. Nature (2020) 587(7832):157–161. doi:10.1038/s41586-020-2833-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, D., Wang, Z., Cupp-Sutton, K. A., Guo, Y., Kou, Q., Smith, K., et al. Quantitative top-down proteomics in complex samples using protein-level tandem mass tag labeling. J. Am. Soc. Mass Spectrom. (2021) 32:1336–1344. doi:10.1021/jasms.0c00464

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H., Gau, B. C., Jones, L. M., Vidavsky, I., and Gross, M. L. Fast photochemical oxidation of proteins for comparing structures of Protein−Ligand complexes: The Calmodulin−Peptide model system. Anal. Chem. (2011) 83(1):311–318. doi:10.1021/ac102426d

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Ruan, C., Zhu, H., Li, K., Zhang, W., Wang, K., et al. A simplified thermal proteome profiling approach to screen protein targets of a ligand. PROTEOMICS (2020) 20(15-16):1900372. doi:10.1002/pmic.201900372

CrossRef Full Text | Google Scholar

Zhao, T., Tian, J., Wang, X., Hao, Y., Xu, M., Zhao, Y., et al. PACTS-assisted thermal proteome profiling for use in identifying peptide-interacting proteins. Anal. Chem. (2022) 94(18):6809–6818. doi:10.1021/acs.analchem.2c00581

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhong, Q., Simonis, N., Li, Q.-R., Charloteaux, B., Heuze, F., Klitgord, N., et al. Edgetic perturbation models of human inherited disorders. Mol. Syst. Biol. (2009) 5(1):321. doi:10.1038/msb.2009.80

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: proteomics, protein thermal stability, proteoform, mass spec, top-down proteomics

Citation: Kang J, Seshadri M, Cupp-Sutton KA and Wu S (2023) Toward the analysis of functional proteoforms using mass spectrometry-based stability proteomics. Front. Anal. Sci. 3:1186623. doi: 10.3389/frans.2023.1186623

Received: 15 March 2023; Accepted: 10 May 2023;
Published: 22 June 2023.

Edited by:

Nicholas B. Borotto, University of Nevada, Reno, United States

Reviewed by:

Michael Fitzgerald, Duke University, United States
André Mateus, Umeå University, Sweden

Copyright © 2023 Kang, Seshadri, Cupp-Sutton and Wu. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Kellye A. Cupp-Sutton, kellyecuppsutton@ou.edu; Si Wu, si.wu@ou.edu

Download