Emerging Proteomic Technologies for Accelerating Development of Senotherapeutics: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Contributor: , , , , , , ,

The accumulation of senescent cells is now widely known to be a driver of aging and many age-related pathologies, such as neurodegeneration and type 2 diabetes, among others. Targeting senescent cells for selective removal or altering the proteins they release are promising therapeutic strategies against age-related diseases. However, the biology of senescent cells is complex, dynamic, and heterogeneous. In order to better identify pathology-driving senescent cells and develop therapies to alter their complex biology or drive them toward cell death, a detailed and comprehensive understanding of senescence-associated proteins and the mechanisms that enable senescent cells to evade cell death is required. Major developments in proteomic workflows over the past decade have enabled an increasingly comprehensive, quantitative, and specific molecular profiling and interrogation of cellular mechanisms.

  • senescence
  • mass spectrometry
  • proteomics
  • biomarkers

1. Introduction

In recent years, proteomics has become an attractive tool for uncovering senescence biology, biomarkers, and therapeutic targets. Researchers anticipate the application of mass spectrometry (MS)-based proteomics will accelerate the translation therapies targeting senescent cells. MS-based proteomics is a gold-standard technique for large-scale protein identification and quantification in complex samples. To overcome the challenges associated with analyzing complex protein samples with a large dynamic range of protein concentrations, sophisticated separation techniques and advanced MS instrumentation have been developed to extend coverage and sensitivity to accommodate the detection of biologically relevant and low-abundance proteins.

2. Emerging MS-Based Approaches for Biomarker Discovery

2.1. Data Acquisition and MS Instrumentation

MS data acquisition methods and analysis pipelines have significantly improved data reproducibility, quantification, and completeness in mass spectrometry-based workflows. The advancement of data-independent acquisition (DIA or SWATH) methods is a major step toward increased reproducibility and quantitative accuracy for biomarker discovery compared with traditional data-dependent acquisition (DDA) methods applied in untargeted proteomics studies [1][2][3]. DIA MS acquires MS/MS spectra for all peptides in a sample without requiring precursor information, which overcomes several challenges associated with DDA, including reduced bias toward abundant peptide selection that results in data missingness and decreased quantitative interference at the MS1 level [2][4]. DIA-MS also offers a reliable and consistent analysis of complex proteomic samples [5][6] that have been validated in consortium-level biomarker studies [3]. DIA analysis also lends itself nicely to PTM profiling and quantification, as quantification at the MS2 level allows one to localize PTM sites and distinguish peptide isomers via site-localized fragment ions [7][8]. DIA-MS acquisition generally uses wider precursor isolation windows to activate all ions for collision in each mass-to-charge (m/z) range, repeated across the full mass range in a single duty cycle. Innovations to the size and placement of precursor isolation windows, such as variable-window DIA [9] and overlapped (staggered)-window DIA [10][11], were developed to improve peptide selectivity. The generation of the first comprehensive SASP profiles in SASP Atlas were generated utilizing variable-window DIA to identify candidate senescence biomarkers such as GDF15, STC1, and SERPINs that are associated with aging in human plasma [12]. Another newly developed novel acquisition method called ‘parallel accumulation–serial fragmentation’ (PASEF) [13], combined with DIA (diaPASEF) [14], improves sensitivity and scanning speed. Unlike typical DIA approaches based predominantly on chromatographic separation, diaPASEF increases the sensitivity and coverage of proteomic analysis with the inclusion of an additional ‘ion mobility’ peptide separation dimension.

2.2. Identification of ‘Proteoform’-Level Biomarkers with Top-Down Proteomics

Hundreds of thousands of proteins are produced by approximately 20,000 human coding genes [15][16][17]. Following protein biosynthesis, post-translational modification (PTM) and proteolytic processing events can modify protein structures and interactions [18][19]. PTMs include chemical groups such as phosphorylation and acetylation and more complex glycan structures, among many others [20][21]. Any protein variation (protein products of a single gene) due to PTMs, alternative RNA splicing, and genetic mutation is defined by a term named ‘proteoform’ [22], which can have distinct functions and change dynamically in response to diverse stimuli [23][24]. Proteoforms are challenging to characterize because of their similar sequences and masses, which make proteomic analysis more complex [15]. Most ‘discovery’ proteomic approaches rely on peptide-level measurements or antibody binding at a specific location on a protein, and therefore are unable to distinguish proteoforms. However, proteoform-level measurements will potentially improve the sensitivity and specificity of proteomic biomarkers.
Top-down proteomics is a powerful technology for analysis of the intact proteins/peptides via tandem mass spectrometry to characterize unique proteoforms and localize PTMs [23][24][25] to reveal structural and functional information on each proteoform [26][27]. Based on recent studies, protein modifications and diseases can be closely associated with each other [28]. For instance, the relationship between the occurrence and development of cancer and phosphorylation sites has been described [29][30][31]. The Kelleher Research Group at Northwestern University has reported 29,620 unique proteoforms which are expressed from human blood and bone marrow, accessible in the Blood Proteoform Atlas (BPA) (https://blood-proteoform-atlas.org (accessed on 24 August 2023)). The potential applications of BPA in clinical validation, such as the prostate-specific antigen isoform (IsoPSA) test in prostate cancer, have been noted [32] and there are many more proteoform associations yet to be explored.
Proteoforms are potential biomarkers that can be directly connected to multifaceted phenotypes [27], and thus the separation of intact proteins is necessary and one of the most significant challenges of top-down proteomics. Intact proteins are typically separated using reversed-phase liquid chromatography (RPLC) coupled with tandem mass spectrometry (MS/MS) analysis. Separation techniques have been developed for multidimensional separation platforms such as capillary zone electrophoresis (CZE), two-dimensional (2D) gel electrophoresis, and 2D-LC platform using high-pH RPLC and low-pH RPLC separation [33][34]. In polyacrylamide gel electrophoresis (PAGE), Passively Eluting Proteins from Polyacrylamide gels as Intact species for MS (‘PEPPI-MS’) is a recent advancement for clean separation and extraction of intact proteins [35]. Future advancements in MS instrumentation and data analysis tools are needed for pervasive application of top-down proteomics in biomedical research [31]. such as streamling and upgrading of database search tools. For example, the presence of unexpected modifications complicates database matching. Fortunately, there is a rapid growth of top-down proteomics [36], propelled by community groups such as the Consortium of Top-Down Proteomics (www.topdownproteomics.org (accessed on 24 August 2023)) and initiatives such as the Human Proteoform Project, which strive to map the human proteoform at proteoform resolution [26].

2.3. Sample Preparation Workflows to Overcome Challenges in Blood Biomarker Discovery

Circulating biomarkers are promising indicators to quantify age-related declines and senescent cell burden [37][38]. The translation of therapies, like senolytics and senomorphics, will require biomarkers of senescence burden to identify and stratify individuals with elevated senescent cell burden and evaluate the efficacy of senolytic interventions. Interrogation of the blood proteome is key to establishing senescence biomarkers and subsequent senotherapeutics. However, the high dynamic range of protein abundance in plasma/serum, which spans over 10 orders of magnitude, poses a challenge in the detection of lower abundance components using mass spectrometry [39]. The 22 most abundant proteins in blood, including albumin, immunoglobulins, myoglobin, transferrin, and haptoglobin, together constitute 99% of the total protein content, whereas albumin itself comprises nearly 60% of the total serum proteome [40]. Low-abundant proteins having biomarker potential may be obscured from detection by high-abundance proteins. Thus, sample preparation protocols are critical for minimizing sample complexity. Various strategies therefore play a crucial role in reducing sample complexity before LC-MS/MS analysis, including depletion of high-abundance proteins, affinity enrichment of low-abundant proteins, and a variety of chromatographic [41] and electrophoretic fractionation techniques [42]. The depletion of highly abundant proteins, reviewed in detail elsewhere [43], is a viable strategy for reducing the dynamic range at the expense of increasing the number of samples to be analyzed. Immunodepleting highly abundant proteins is a routinely applied method with high specificity and reproducibility [43]. Antibody-based depletion of the top 7 or top 14 high-abundance plasma proteins showed 25% gains in proteome coverage [44]. However, these gains come at the expense of added processing steps, additional variability, and the cost of the depletion columns. Another method based on bead-bound hexapeptides known as combinatorial peptide ligand library (CPLL, commercially named Proteominer) reduces the dynamic range of protein concentrations [45]. CPLL enriches low-abundant proteins in a reproducible manner [46], and with greater performance compared to the other available depletion/enrichment approaches, yielding 68.6% higher protein identification compared to the untreated serum [47]. The impact of CPLL in serum/plasma biomarker discovery is excellently reviewed elsewhere [48]. In one notable study, CPLL increased plasma protein identifications by 22% in breast cancer patients, of which 23 were differentially expressed and thus potential biomarker candidates [49]. Another study applying CPLL identified 26 differentially expressed proteins, of which ficolin-2 was reported as a potential diagnostic marker for rheumatoid arthritis [50].
Each strategy employed for reducing the dynamic range of protein samples has its own merits and drawbacks [43][51] that impose trade-offs between sample throughput and proteome coverage. Extensive offline fractionation increases the number of samples and MS analysis time, whereas depletion/enrichment strategies add processing steps that introduce technical variability and cost. Further depth can be achieved by utilizing a combination of approaches [52][53]. A recent study assessed the plasma proteome by comparing depletion via Human 6 (Hu6) and Human 14 (Hu14) columns and the ProteoMiner Kit, each in combination with protein level fractionation via SDS PAGE or via offline C18 chromatography before trypsin digestion [54]. This study identified a total of 4385 proteins, 3064 (70%) of which were common between all methods. Another study reported 3400 proteins from pooled plasma samples from AD patients and healthy controls [55] by using a combination of immune depletion, HPLC fractionation, and 2D gel electrophoresis before LC-MS/MS analysis. Throughput loss resulting from extensive fractionation can be mitigated by the utilization of isobaric tags for relative and absolute quantification (iTRAQ) or tandem mass tags (TMT) for multiplexing analysis [56]. Keshishian et al. utilized abundant protein depletion, iTRAQ isobaric labeling, and offline basic PH reversed-phase fractionation to identify >4500 proteins in plasma with high reproducibility [57].
A recently developed nanoparticle-based method has reported promising results for the analysis of blood samples [58][59]. Over the last decade, there has been an increased understanding of the mechanisms of generation of ‘protein corona’, or the protein mixture bound at the interface between the surface of nanoparticles and a biological fluid, and which factors influence its composition [60][61]. Nanoparticles can reduce the proportion of highly abundant proteins and enrich low-abundance proteins in serum and plasma. Notably, the composition of protein coronas can be tuned for a swath of plasma proteins in a reproducible manner, enabling the use of nanoparticle-based automation workflows in combination with MS analysis, enabling protein coverage exceeding 2000 proteins [62]. Although initial studies utilizing nanoparticles suggest great promise for proteomic analysis of blood samples, these methods need to be further evaluated and improve reproducibility across different cores and institutions [63]. Improvements in standardization and automation will aid in these efforts [58].

3. MS-Based Approaches for the Discovery of Novel Therapeutic Targets

Proteomic methodologies can be leveraged for the discovery of novel senotherapeutic approaches in multiple ways. First, the identification of senescence-specific cell-surface proteins will unveil senotherapeutic targets. Second, proteomic profiling of protein–drug interactions associated with senolysis can aid in the identification of new pathways and proteins for the development of senolytics. As the protein targets of several existing senolytics are not yet known, the identification of protein interactors from existing senolytics presents an intriguing area for the discovery of novel senolytic pathways and the development of new drugs. Identifying specific drug/protein binding interactions may also enable the development of scientific interventions to address issues such as drug delivery, bioavailability, and off-target binding. Numerous methods for proteomic target discovery have surfaced over the last few decades, many of them relying on the quantitative unbiased measurements given by mass spectrometry.
Several existing methods, Thermal Proteome Profiling (TPP), Limited Proteolysis (LIP), and Stability of Proteins from Rates of Oxidation (SPROX), have proven to be effective in the successful identification and quantification of specific intracellular drug/target binding interactions. New approaches, in addition to these previously well-established methods, have also been recently introduced such as pulse proteolysis (PP) and Proteome Integral Solubility Alteration (PISA).

3.1. Limited Proteolysis Coupled to Mass Spectrometry (LiP-MS)

Lip-MS is a technique that compares proteolytic fragmentation patterns between a drug-treated group and a non-treated control [64]. Drug treatment induces conformational changes in drug-bound protein targets. Then, when a broad-spectrum protease is added to the samples for a short time, these conformational changes cause the protease to have altered accessibility to its cleavage sites at the site of drug–protein interactions. This results in distinct peptide fragment signatures in the drug-treated group compared to the control [64]. After disrupting the protein–drug interaction, specific digestion with trypsin is then used to further break down the remaining large peptide fragments, leaving tryptic peptides enriched at the sites of protein–drug interaction to be detected via MS. Lip-MS is useful in complex mixtures, such as cell lysates, for understanding the structure of proteins and any changes that occur due to drug binding [65].

3.2. Pulse Proteolysis (PP)

Pulse proteolysis is another proteomic technique used for identifying potential drug targets, as well as protein–protein interactions. In this method, a cell lysate is incubated with a drug and urea, while a control sample is incubated under identical conditions without the drug. The drug will theoretically stabilize its target proteins and prevent their denaturation by urea. Then, a protease is introduced for a short period, referred to as a ‘pulse’. PP causes complete digestion of unfolded proteins, but drug-stabilized proteins remain intact [66]. After the pulse period, the reaction is quenched by rapidly lowering the temperature or by adding protease inhibitors. The protein is run on a gel, and the intact protein bands (higher molecular weight) are quantitatively compared. Drug-bound targets are indicated by an increased amount of intact protein in the drug-treated group [66]. A later improvement in this method incorporated mass spectrometry for the untargeted discovery of drug-binding interactions. In this variation, the portion of the gel containing intact protein is excised, digested in gel with trypsin, and analyzed via MS to compare the fraction of intact proteins between drug-treated and untreated groups [67].

3.3. Stability of Proteins from Rates of Oxidation (SPROX)

Stability of Proteins from Rates of Oxidation (SPROX) employs a hydrogen peroxide buffer with increasing levels of a denaturant to generate chemical denaturation curves [68]. After an equal amount of time for the oxidation rate, the samples are quenched. Chemical denaturation curves are generated based on the oxidation of methionine residues on peptides detected and quantified with mass spectrometry [68]. SPROX reveals proteins that are stabilized or destabilized by a bound target molecule, indicated by either a right or left shift in the chemical denaturation curve [69]. It is especially beneficial in evaluating the disassociation constants of protein–ligand complexes as oxidation decreases protein stability. Since its first use, SPROX has been updated to allow for higher throughput detection with isobaric tags [69].

3.4. Thermal Denaturation-Based Target Discovery: TPP, CETSA, and PISA

The Cellular Thermal Shift Assay (CETSA) compares thermal denaturation curves between drug-treated and control groups to identify potential drug targets. Drug-bound proteins exhibit stabilization that protects the drug–protein complex from thermal denaturation, allowing for the interaction to be identified by identifying proteins with a right-shifted curve [70]. Thermal Proteome Profiling (TPP) combines CETSA with mass spectrometry analysis to identify novel protein–drug interactions in untargeted mass spectrometry analysis [71]. Both techniques can be used in cell extracts and intact cells, enabling the identification of target engagement within the intact cellular environment. Notably, TPP can also be used to identify drug targets in tissues from animal studies. For example, one study demonstrated the dose-dependent engagement of methionine aminopeptidase-2 (MetAP2) with its inhibitor, TNP-470, in the livers of mice treated with the drug [70]. These techniques are beneficial for target discovery following treatment with a ligand, and TPP can also be applied to analyze downstream effects and other factors that affect protein stability such as post-translational modifications (PTMs) [71]. Finally, Proteome Integral Solubility Alteration (PISA) pairs thermal denaturation with the use of isobaric labeling to enable sample multiplexing during MS analysis [72]. By combining the soluble protein fractions obtained after a thermal gradient, targets can be identified by looking at the area under the curve across plexes within each sample [72]. PISA also allows for comparative quantification at a 10-or-more-fold increased throughput when compared to CETSA/TPP.

3.5. Size-Exclusion Chromatography and Affinity Selection Mass Spectrometry (SEC-ASMS)

SEC-ASMS is a hybrid analytical technique that combines the size separation capability of SEC with further separation via affinity selection which is then analyzed with MS [73][74]. SEC is a liquid chromatography method that separates molecules based on size and shape. Affinity selection allows for the separation of ligand-bound proteins in a complex mixture. After the SEC separation, the eluted protein fractions are subjected to affinity selection, adding the ligand of interest, to selectively capture and enrich their target molecules from the complex protein mixture [73][74]. After affinity selection, captured protein–ligand complexes are analyzed using mass spectrometry to characterize the interaction and identify targets [73][74]. SEC-ASMS is also helpful for examining protein complexes, protein–protein interactions, and mapping post-translational modifications to study their function.

3.6. Targeting and Quantifying Senescent Cells through the ‘Surfaceome’

For decades there have been efforts to identify and quantity proteins on cell surfaces because of their value as markers of cell identity and as therapeutic targets. A prime example of this is immunophenotyping, a method that essentially exploits the presence or absence of specific cell-surface markers to count and discriminate cell populations using antibody detection. Immunophenotyping is now a tool of choice to probe the phenotype and function of the immune system [75]. This method can be used to count and discriminate specific lymphocyte populations, including progenitor cells and various hematopoietic lineages, with ever-increasing granularity based on the presence or absence of cell-surface markers [75][76]. Knowing the identity and quantity of proteins on the cell surface has enormous potential benefits for translational research and the development of disease diagnostics and therapeutics, not only for its established benefit in immunophenotyping, but for the potential to identify therapeutic drugs, immunotherapy targets, and cell-surface targets for the isolation of cell populations.
Mass spectrometry-based proteomics has been applied to a variety of senescence models to identify surface markers that can be used to target these cells for senotherapy (Table 1). For example, unbiased mass spectrometry-based studies have identified dipeptidyl peptidase 4 (DPP4) to be enriched on the surface of senescent versus proliferating fibroblasts [77]. Importantly, DPP4 can be exploited for both sorting senescent cells via flow cytometry and for targeting them in vitro using antibody-dependent cell-mediated cytotoxicity. A subsequent study demonstrated that senescence-associated DPP4 activity can be pharmacologically targeted to improve hemostasis and plaque stability in old mice [78]. Another promising example of a therapeutically relevant surface marker is the urokinase-type plasminogen activator receptor (uPAR), originally identified as a preferentially expressed marker on the surface of senescent human lung adenocarcinoma cells and melanocytes [79]. In the same study, the authors demonstrated that chimeric antigen receptor (CAR) T cells that target uPAR efficiently eliminate senescent cells in vivo and extend the survival of mice with lung adenocarcinoma following treatment with MEK and CDK4/6 inhibitors, and ameliorated liver fibrosis in two independent models [79]. These examples highlight the utility of cell-surface proteins as potential senotherapeutic targets.
Table 1. Surface markers from senescent cells identified using mass spectrometry-based proteomics. RS = replicative senescence; OIS = oncogene-induced senescence; IR = irradiation; PM = plasma membrane; ADCs = antibody–drug conjugates; ADCC = antibody-dependent cell-mediated toxicity.
Surface
Protein
Cell/Tissue Type Senescence Inducer Strategy Senolytic
Validation
Reference
DEP1/PTPRJ/CD148 Bladder cancer (EJp21 and EJp16);
human lung fibroblast (IMR-90);
human fibrosarcoma (HT1080p21);
mouse lung adenomas (V600EBRAF);
human melanocytes
p21/p16 overexpression;
RS, RAS OIS
-
-
-
PM isolation, in-gel digestion, and LC-MS/MS   [80]
B2MG Bladder cancer (EJp21 and EJp16);
mouse lung adenomas (V600EBRAF)
p21/p16 overexpression
-
PM isolation, in-gel digestion, and LC-MS/MS Gold nanoparticles, ADCs [80][81][82]
NOTCH1 Human lung fibroblast (IMR-90);
mouse pancreatic neoplasm (p48-cre)
HRASG12V, OIS, Etoposide
KrasG12D OIS
SILAC   [83]
DPP4/CD26 Human lung fibroblast (WI-38, IMR-90);
human aortic endothelial cells (HAEC);
human umbilical vein endothelial cells (HUVEC);
mouse embryonic fibroblasts (MEF)
RS, IR, Doxorubicin
IR
IR

HRASG12V OIS
PM isolation, in-gel digestion, and LC-MS/MS ADCC [77]
SCAMP4 Human lung fibroblast (WI-38, IMR-90);
human aortic endothelial cells (HAEC);
human umbilical vein endothelial cells (HUVEC)
RS, IR, Doxorubicin, HRASG12V OIS
IR
IR
PM isolation, in-gel digestion, and LC-MS/MS   [84]
CD24 Bone marrow mesenchymal cells from INK-ATTAC mice - CyTOF   [85]
Despite its huge benefits in diagnostics and cell phenotyping, immunophenotyping is a limited tool for the discovery of new cell-surface markers. First, it is limited to the set of cell-surface proteins that are already known and have high-quality antibodies. Secondly, immunophenotyping has limited multiplexing capability. Thus, unbiased approaches are critically needed for the discovery of new surfaceome targets that will aid in the discrimination of the highly diverse and heterogeneous cell types in the human body. Technological advancements in mass spectrometry approaches now enable comprehensive and unbiased identification and quantification of surfaceome proteins. Over the past decade, an increasing number of sample preparation workflows, computational tools, and curated surfaceomes have emerged to aid in the discovery and validation of cell-surface proteins using mass spectrometry, and are reviewed more extensively elsewhere [86][87]. The most rigorous methods for identification of the cell-surface proteome and the surface interactome utilize biorthogonal chemistry to biotinylate cell-surface proteins for subsequent affinity enrichment. For example, the cell-surface capture (CSC) [88] method utilizes non-cell-permeable reagents to biotinylate cell-surface glycoproteins, which comprise 90% of cell-surface proteins. Following the labeling of live cells in culture, labeled glycoproteins can be enriched either at the protein or peptide levels before mass spectrometry analysis. Modifications of CSC include Cys-Glyco-CSC, which allows the co-enrichment of peptides that are linked to the glycopeptides via disulfide bonds, and Lys-CSC, which allows labeling and enrichment of all surface-exposed lysine-containing proteins [89]. Modern variations of CSC have enhanced its utility for smaller sample inputs [90] and implemented automation, such as autoCSC [91][92] and µCSC [93], resulting in vastly improved sensitivity, reproducibility, and throughput and making surfaceome characterizations of many samples and cell types more feasible. Another recent innovation termed LUX-MS enables a precise spatiotemporal elucidation of the surface interactome of ligands or antibodies using a light-mediated reaction, thus enabling the precise mapping of cell-surface nanoscale organization with targeted therapies [94].
A critical aspect of mass spectrometry-based identification of the surfaceome is the correct annotation of surface proteins from the huge proteomic dataset, which requires a stringent bioinformatic analysis based on specific characteristics of surface proteins. A widely used approach is gene ontology (GO) annotations to identify which proteins are reported to be associated with the cell surface or plasma membrane. More rigorous filtering criteria for the identification of surface glycoproteins after CSC rely on the detection of a mass shift of 0.984 Da that corresponds to the deamidation of asparagine residues through enzymatic deglycosylation during the enrichment process. The selected peptides can be further filtered for the presence of the conserved NXS/T motif that is present in more than 90% of the N-glycosylation sites, allowing the exclusion of the peptides that might have co-eluted during the enrichment process [91]. Similarly, Lys-CSC requires the selection of all peptides that contain a mass shift of 145.019 Da corresponding to the 3-(carbamidomethylthio) propanoyl modification on the lysine residues [89].
Numerous software tools and databases can be employed to aid in the identification, filtering, and prioritization of candidate cell-surface proteins identified from mass spectrometry-based approaches. A suite of tools in the CellSurfer portal [93] (https://www.cellsurfer.net/ (accessed on 29 September 2023)), developed by the Gundry laboratory, is one such resource that aids in annotating and scoring candidate cell-surface markers from mass spectrometry data. The site includes several tools [93][95][96] that enable the rigorous bioinformatic validation, prioritization, ranking, and visualization of candidate surface proteins for future validation and clinical studies. Protter (https://wlab.ethz.ch/protter/start/ (accessed on 29 September 2023)) is another online open-source tool that allows visualization of peptides in the protein topology and provides evidence to validate whether the identified peptides are exposed on the cell surface [97].
Increased interest in exploring the cell surfaceome as a source of potential biomarkers has led to the development of online databases that can serve as a reference for future surfaceome studies. The Cell Surface Protein Atlas [98] (https://wlab.ethz.ch/cspa/ (accessed on 29 September 2023)) is a repository of mass spectrometry-derived cell-surface proteins from 41 human and 31 mouse cell types, providing a snapshot of the surface proteome from different cell types, including cancer cells. Similarly, the in silico human surfaceome (http://wlab.ethz.ch/surfaceome/ accessed on 29 September 2023)) is an online database containing 2886 proteins predicted using a machine-learning-based predictor, SURFY, developed by the Wollschied laboratory [99]. Despite these available databases, the unique cell surfaceome signature has not yet been described for many cell types and disease states, and thus continued efforts and innovation in surface protein discovery and annotation are needed.
There are also potential limitations to utilizing the surfaceome for targeting. First, just as in other markers, cell-surface markers are heterogeneous on senescent cells. Even in presumably homogenous cell culture conditions, not all induced senescent cells express surface markers such as DPP4 [77]. This is particularly true for markers of senescence, since senescent cell populations are inherently heterogenous and markers identified via bulk proteomic approaches may overestimate the ability of a protein to be a true marker of senescence. Strategies coupling single-cell technologies with surface labeling can provide a better approach to identifying markers from such heterogenous populations [86][87]. Secondly, it is not yet known how well surface markers distinguish different populations of senescent cells based on their phenotypes. Another technical challenge is that the validation of surface markers discovered via MS is limited by the availability of highly sensitive and specific antibodies to cell-surface proteins. This may limit the validation of surfaceome candidates using orthogonal approaches such as flow cytometry.

This entry is adapted from the peer-reviewed paper 10.3390/biology12101301

References

  1. Neri, F.; Basisty, N.; Desprez, P.Y.; Campisi, J.; Schilling, B. Quantitative Proteomic Analysis of the Senescence-Associated Secretory Phenotype by Data-Independent Acquisition. Curr. Protoc. 2021, 1, e32.
  2. Gillet, L.C.; Navarro, P.; Tate, S.; Rost, H.; Selevsek, N.; Reiter, L.; Bonner, R.; Aebersold, R. Targeted data extraction of the MS/MS spectra generated by data-independent acquisition: A new concept for consistent and accurate proteome analysis. Mol. Cell. Proteom. 2012, 11, O111.016717.
  3. Collins, B.C.; Hunter, C.L.; Liu, Y.; Schilling, B.; Rosenberger, G.; Bader, S.L.; Chan, D.W.; Gibson, B.W.; Gingras, A.C.; Held, J.M.; et al. Multi-laboratory assessment of reproducibility, qualitative and quantitative performance of SWATH-mass spectrometry. Nat. Commun. 2017, 8, 291.
  4. Sajic, T.; Liu, Y.; Aebersold, R. Using data-independent, high-resolution mass spectrometry in protein biomarker research: Perspectives and clinical applications. Proteom. Clin. Appl. 2015, 9, 307–321.
  5. Dey, A.K.; Kumar, B.; Singh, A.K.; Ranjan, P.; Thiruvengadam, R.; Desiraju, B.K.; Kshetrapal, P.; Wadhwa, N.; Bhatnagar, S.; Rashid, F.; et al. Salivary proteome signatures in the early and middle stages of human pregnancy with term birth outcome. Sci. Rep. 2020, 10, 8022.
  6. Kumar, B.; Dey, A.K.; Saha, S.; Singh, A.K.; Kshetrapal, P.; Wadhwa, N.; Thiruvengadam, R.; Desiraju, B.K.; Bhatnagar, S.; Salunke, D.M.; et al. Dynamic Alteration in the Vaginal Secretory Proteome across the Early and Mid-Trimesters of Pregnancy. J. Proteome Res. 2021, 20, 1190–1205.
  7. Xie, X.; Shah, S.; Holtz, A.; Rose, J.; Basisty, N.; Schilling, B. Simultaneous Affinity Enrichment of Two Post-Translational Modifications for Quantification and Site Localization. J. Vis. Exp. 2020, 156, e60780.
  8. Basisty, N.; Meyer, J.G.; Wei, L.; Gibson, B.W.; Schilling, B. Simultaneous Quantification of the Acetylome and Succinylome by ‘One-Pot’ Affinity Enrichment. Proteomics 2018, 18, e1800123.
  9. Zhang, Y.; Bilbao, A.; Bruderer, T.; Luban, J.; Strambio-De-Castillia, C.; Lisacek, F.; Hopfgartner, G.; Varesio, E. The Use of Variable Q1 Isolation Windows Improves Selectivity in LC-SWATH-MS Acquisition. J. Proteome Res. 2015, 14, 4359–4371.
  10. Amodei, D.; Egertson, J.; MacLean, B.X.; Johnson, R.; Merrihew, G.E.; Keller, A.; Marsh, D.; Vitek, O.; Mallick, P.; MacCoss, M.J. Improving Precursor Selectivity in Data-Independent Acquisition Using Overlapping Windows. J. Am. Soc. Mass Spectrom. 2019, 30, 669–684.
  11. Pino, L.K.; Just, S.C.; MacCoss, M.J.; Searle, B.C. Acquiring and Analyzing Data Independent Acquisition Proteomics Experiments without Spectrum Libraries. Mol. Cell. Proteom. 2020, 19, 1088–1103.
  12. Basisty, N.; Kale, A.; Jeon, O.H.; Kuehnemann, C.; Payne, T.; Rao, C.; Holtz, A.; Shah, S.; Sharma, V.; Ferrucci, L.; et al. A proteomic atlas of senescence-associated secretomes for aging biomarker development. PLoS Biol. 2020, 18, e3000599.
  13. Meier, F.; Brunner, A.D.; Koch, S.; Koch, H.; Lubeck, M.; Krause, M.; Goedecke, N.; Decker, J.; Kosinski, T.; Park, M.A.; et al. Online Parallel Accumulation-Serial Fragmentation (PASEF) with a Novel Trapped Ion Mobility Mass Spectrometer. Mol. Cell. Proteom. 2018, 17, 2534–2545.
  14. Meier, F.; Brunner, A.D.; Frank, M.; Ha, A.; Bludau, I.; Voytik, E.; Kaspar-Schoenefeld, S.; Lubeck, M.; Raether, O.; Bache, N.; et al. diaPASEF: Parallel accumulation-serial fragmentation combined with data-independent acquisition. Nat. Methods 2020, 17, 1229–1236.
  15. Walker, K.A.; Basisty, N.; Wilson, D.M., 3rd; Ferrucci, L. Connecting aging biology and inflammation in the omics era. J. Clin. Investig. 2022, 132, e158448.
  16. Rifai, N.; Gillette, M.A.; Carr, S.A. Protein biomarker discovery and validation: The long and uncertain path to clinical utility. Nat. Biotechnol. 2006, 24, 971–983.
  17. Doll, S.; Burlingame, A.L. Mass spectrometry-based detection and assignment of protein posttranslational modifications. ACS Chem. Biol. 2015, 10, 63–71.
  18. Olzscha, H. Posttranslational modifications and proteinopathies: How guardians of the proteome are defeated. Biol. Chem. 2019, 400, 895–915.
  19. Retanal, C.; Ball, B.; Geddes-McAlister, J. Post-Translational Modifications Drive Success and Failure of Fungal-Host Interactions. J. Fungi 2021, 7, 124.
  20. Ramazi, S.; Zahiri, J. Posttranslational modifications in proteins: Resources, tools and prediction methods. Database 2021, 2021, baab012.
  21. Chen, L.; Kashina, A. Post-translational Modifications of the Protein Termini. Front. Cell Dev. Biol. 2021, 9, 719590.
  22. Smith, L.M.; Kelleher, N.L.; Linial, M.; Goodlett, D.; Langridge-Smith, P.; Ah Goo, Y.; Safford, G.; Bonilla, L.; Kruppa, G.; Zubarev, R.; et al. Proteoform: A single term describing protein complexity. Nat. Methods 2013, 10, 186–187.
  23. Holtz, A.; Basisty, N.; Schilling, B. Quantification and Identification of Post-Translational Modifications Using Modern Proteomics Approaches. Methods Mol. Biol. 2021, 2228, 225–235.
  24. Smith, L.M.; Kelleher, N.L. Proteoforms as the next proteomics currency. Science 2018, 359, 1106–1107.
  25. Brodbelt, J.S. Deciphering combinatorial post-translational modifications by top-down mass spectrometry. Curr. Opin. Chem. Biol. 2022, 70, 102180.
  26. Melby, J.A.; Roberts, D.S.; Larson, E.J.; Brown, K.A.; Bayne, E.F.; Jin, S.; Ge, Y. Novel Strategies to Address the Challenges in Top-Down Proteomics. J. Am. Soc. Mass Spectrom. 2021, 32, 1278–1294.
  27. Toby, T.K.; Fornelli, L.; Kelleher, N.L. Progress in Top-Down Proteomics and the Analysis of Proteoforms. Annu. Rev. Anal. Chem. 2016, 9, 499–519.
  28. Zhong, Q.; Xiao, X.; Qiu, Y.; Xu, Z.; Chen, C.; Chong, B.; Zhao, X.; Hai, S.; Li, S.; An, Z.; et al. Protein posttranslational modifications in health and diseases: Functions, regulatory mechanisms, and therapeutic implications. MedComm 2023, 4, e261.
  29. Tong, Y.; Sun, M.; Chen, L.; Wang, Y.; Li, Y.; Li, L.; Zhang, X.; Cai, Y.; Qie, J.; Pang, Y.; et al. Proteogenomic insights into the biology and treatment of pancreatic ductal adenocarcinoma. J. Hematol. Oncol. 2022, 15, 168.
  30. Krug, K.; Jaehnig, E.J.; Satpathy, S.; Blumenberg, L.; Karpova, A.; Anurag, M.; Miles, G.; Mertins, P.; Geffen, Y.; Tang, L.C.; et al. Proteogenomic Landscape of Breast Cancer Tumorigenesis and Targeted Therapy. Cell 2020, 183, 1436–1456.e31.
  31. Satpathy, S.; Krug, K.; Jean Beltran, P.M.; Savage, S.R.; Petralia, F.; Kumar-Sinha, C.; Dou, Y.; Reva, B.; Kane, M.H.; Avanessian, S.C.; et al. A proteogenomic portrait of lung squamous cell carcinoma. Cell 2021, 184, 4348–4371.e40.
  32. Melani, R.D.; Gerbasi, V.R.; Anderson, L.C.; Sikora, J.W.; Toby, T.K.; Hutton, J.E.; Butcher, D.S.; Negrão, F.; Seckler, H.S.; Srzentić, K.; et al. The Blood Proteoform Atlas: A reference map of proteoforms in human hematopoietic cells. Science 2022, 375, 411–418.
  33. Wang, Z.; Yu, D.; Cupp-Sutton, K.A.; Liu, X.; Smith, K.; Wu, S. Development of an Online 2D Ultrahigh-Pressure Nano-LC System for High-pH and Low-pH Reversed Phase Separation in Top-Down Proteomics. Anal. Chem. 2020, 92, 12774–12777.
  34. McCool, E.N.; Lubeckyj, R.A.; Shen, X.; Chen, D.; Kou, Q.; Liu, X.; Sun, L. Deep Top-Down Proteomics Using Capillary Zone Electrophoresis-Tandem Mass Spectrometry: Identification of 5700 Proteoforms from the Escherichia coli Proteome. Anal. Chem. 2018, 90, 5529–5533.
  35. Takemori, A.; Butcher, D.S.; Harman, V.M.; Brownridge, P.; Shima, K.; Higo, D.; Ishizaki, J.; Hasegawa, H.; Suzuki, J.; Yamashita, M.; et al. PEPPI-MS: Polyacrylamide-Gel-Based Prefractionation for Analysis of Intact Proteoforms and Protein Complexes by Mass Spectrometry. J. Proteome Res. 2020, 19, 3779–3791.
  36. He, L.; Rockwood, A.L.; Agarwal, A.M.; Anderson, L.C.; Weisbrod, C.R.; Hendrickson, C.L.; Marshall, A.G. Top-down proteomics-a near-future technique for clinical diagnosis? Ann. Transl. Med. 2020, 8, 136.
  37. Basisty, N.; Kale, A.; Patel, S.; Campisi, J.; Schilling, B. The power of proteomics to monitor senescence-associated secretory phenotypes and beyond: Toward clinical applications. Expert Rev. Proteom. 2020, 17, 297–308.
  38. Moaddel, R.; Ubaida-Mohien, C.; Tanaka, T.; Lyashkov, A.; Basisty, N.; Schilling, B.; Semba, R.D.; Franceschi, C.; Gorospe, M.; Ferrucci, L. Proteomics in aging research: A roadmap to clinical, translational research. Aging Cell 2021, 20, e13325.
  39. Hortin, G.L.; Sviridov, D. The dynamic range problem in the analysis of the plasma proteome. J. Proteom. 2010, 73, 629–636.
  40. Zhang, H.; Yi, E.C.; Li, X.J.; Mallick, P.; Kelly-Spratt, K.S.; Masselon, C.D.; Camp, D.G., 2nd; Smith, R.D.; Kemp, C.J.; Aebersold, R. High throughput quantitative analysis of serum proteins using glycopeptide capture and liquid chromatography mass spectrometry. Mol. Cell. Proteom. 2005, 4, 144–155.
  41. Neverova, I.; Van Eyk, J.E. Role of chromatographic techniques in proteomic analysis. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 2005, 815, 51–63.
  42. Matt, P.; Fu, Z.; Fu, Q.; Van Eyk, J.E. Biomarker discovery: Proteome fractionation and separation in biological samples. Physiol. Genom. 2008, 33, 12–17.
  43. Lee, P.Y.; Osman, J.; Low, T.Y.; Jamal, R. Plasma/serum proteomics: Depletion strategies for reducing high-abundance proteins for biomarker discovery. Bioanalysis 2019, 11, 1799–1812.
  44. Tu, C.; Rudnick, P.A.; Martinez, M.Y.; Cheek, K.L.; Stein, S.E.; Slebos, R.J.; Liebler, D.C. Depletion of abundant plasma proteins and limitations of plasma proteomics. J. Proteome Res. 2010, 9, 4982–4991.
  45. Guerrier, L.; Righetti, P.G.; Boschetti, E. Reduction of dynamic protein concentration range of biological extracts for the discovery of low-abundance proteins by means of hexapeptide ligand library. Nat. Protoc. 2008, 3, 883–890.
  46. Leger, T.; Lavigne, D.; Le Caer, J.P.; Guerrier, L.; Boschetti, E.; Fareh, J.; Feldman, L.; Laprevote, O.; Meilhac, O. Solid-phase hexapeptide ligand libraries open up new perspectives in the discovery of biomarkers in human plasma. Clin. Chim. Acta 2011, 412, 740–747.
  47. Pisanu, S.; Biosa, G.; Carcangiu, L.; Uzzau, S.; Pagnozzi, D. Comparative evaluation of seven commercial products for human serum enrichment/depletion by shotgun proteomics. Talanta 2018, 185, 213–220.
  48. Boschetti, E.; Righetti, P.G. Low-Abundance Protein Enrichment for Medical Applications: The Involvement of Combinatorial Peptide Library Technique. Int. J. Mol. Sci. 2023, 24, 329.
  49. Meng, R.; Gormley, M.; Bhat, V.B.; Rosenberg, A.; Quong, A.A. Low abundance protein enrichment for discovery of candidate plasma protein biomarkers for early detection of breast cancer. J. Proteom. 2011, 75, 366–374.
  50. Cheng, Y.; Chen, Y.; Sun, X.; Li, Y.; Huang, C.; Deng, H.; Li, Z. Identification of potential serum biomarkers for rheumatoid arthritis by high-resolution quantitative proteomic analysis. Inflammation 2014, 37, 1459–1467.
  51. Cao, Z.; Tang, H.Y.; Wang, H.; Liu, Q.; Speicher, D.W. Systematic comparison of fractionation methods for in-depth analysis of plasma proteomes. J. Proteome Res. 2012, 11, 3090–3100.
  52. Mauri, P.; Scigelova, M. Multidimensional protein identification technology for clinical proteomic analysis. Clin. Chem. Lab. Med. 2009, 47, 636–646.
  53. Geyer, P.E.; Holdt, L.M.; Teupser, D.; Mann, M. Revisiting biomarker discovery by plasma proteomics. Mol. Syst. Biol. 2017, 13, 942.
  54. Kaur, G.; Poljak, A.; Ali, S.A.; Zhong, L.; Raftery, M.J.; Sachdev, P. Extending the Depth of Human Plasma Proteome Coverage Using Simple Fractionation Techniques. J. Proteome Res. 2021, 20, 1261–1279.
  55. Laffoon, S.B.; Doecke, J.D.; Roberts, A.M.; Vance, J.A.; Reeves, B.D.; Pertile, K.K.; Rumble, R.L.; Fowler, C.J.; Trounson, B.; Ames, D.; et al. Analysis of plasma proteins using 2D gels and novel fluorescent probes: In search of blood based biomarkers for Alzheimer’s disease. Proteome Sci. 2022, 20, 2.
  56. Moulder, R.; Bhosale, S.D.; Goodlett, D.R.; Lahesmaa, R. Analysis of the plasma proteome using iTRAQ and TMT-based Isobaric labeling. Mass Spectrom. Rev. 2018, 37, 583–606.
  57. Keshishian, H.; Burgess, M.W.; Specht, H.; Wallace, L.; Clauser, K.R.; Gillette, M.A.; Carr, S.A. Quantitative, multiplexed workflow for deep analysis of human blood plasma and biomarker discovery by mass spectrometry. Nat. Protoc. 2017, 12, 1683–1701.
  58. Ferdosi, S.; Tangeysh, B.; Brown, T.R.; Everley, P.A.; Figa, M.; McLean, M.; Elgierari, E.M.; Zhao, X.; Garcia, V.J.; Wang, T.; et al. Engineered nanoparticles enable deep proteomics studies at scale by leveraging tunable nano–bio interactions. Proc. Natl. Acad. Sci. USA 2022, 119, e2106053119.
  59. Meng, Y.; Chen, J.; Liu, Y.; Zhu, Y.; Wong, Y.K.; Lyu, H.; Shi, Q.; Xia, F.; Gu, L.; Zhang, X.; et al. A highly efficient protein corona-based proteomic analysis strategy for the discovery of pharmacodynamic biomarkers. J. Pharm. Anal. 2022, 12, 879–888.
  60. Tenzer, S.; Docter, D.; Rosfa, S.; Wlodarski, A.; Kuharev, J.; Rekik, A.; Knauer, S.K.; Bantz, C.; Nawroth, T.; Bier, C.; et al. Nanoparticle size is a critical physicochemical determinant of the human blood plasma corona: A comprehensive quantitative proteomic analysis. ACS Nano 2011, 5, 7155–7167.
  61. Bertrand, N.; Grenier, P.; Mahmoudi, M.; Lima, E.M.; Appel, E.A.; Dormont, F.; Lim, J.M.; Karnik, R.; Langer, R.; Farokhzad, O.C. Mechanistic understanding of in vivo protein corona formation on polymeric nanoparticles and impact on pharmacokinetics. Nat. Commun. 2017, 8, 777.
  62. Blume, J.E.; Manning, W.C.; Troiano, G.; Hornburg, D.; Figa, M.; Hesterberg, L.; Platt, T.L.; Zhao, X.; Cuaresma, R.A.; Everley, P.A.; et al. Rapid, deep and precise profiling of the plasma proteome with multi-nanoparticle protein corona. Nat. Commun. 2020, 11, 3662.
  63. Ashkarran, A.A.; Gharibi, H.; Voke, E.; Landry, M.P.; Saei, A.A.; Mahmoudi, M. Measurements of heterogeneity in proteomics analysis of the nanoparticle protein corona across core facilities. Nat. Commun. 2022, 13, 6610.
  64. Schopper, S.; Kahraman, A.; Leuenberger, P.; Feng, Y.; Piazza, I.; Muller, O.; Boersema, P.J.; Picotti, P. Measuring protein structural changes on a proteome-wide scale using limited proteolysis-coupled mass spectrometry. Nat. Protoc. 2017, 12, 2391–2410.
  65. Pepelnjak, M.; de Souza, N.; Picotti, P. Detecting Protein-Small Molecule Interactions Using Limited Proteolysis-Mass Spectrometry (LiP-MS). Trends Biochem. Sci. 2020, 45, 919–920.
  66. Park, C.; Marqusee, S. Pulse proteolysis: A simple method for quantitative determination of protein stability and ligand binding. Nat. Methods 2005, 2, 207–212.
  67. Adhikari, J.; Fitzgerald, M.C. SILAC-pulse proteolysis: A mass spectrometry-based method for discovery and cross-validation in proteome-wide studies of ligand binding. J. Am. Soc. Mass Spectrom. 2014, 25, 2073–2083.
  68. West, G.M.; Tang, L.; Fitzgerald, M.C. Thermodynamic analysis of protein stability and ligand binding using a chemical modification- and mass spectrometry-based strategy. Anal. Chem. 2008, 80, 4175–4185.
  69. Strickland, E.C.; Geer, M.A.; Tran, D.T.; Adhikari, J.; West, G.M.; DeArmond, P.D.; Xu, Y.; Fitzgerald, M.C. Thermodynamic analysis of protein-ligand binding interactions in complex biological mixtures using the stability of proteins from rates of oxidation. Nat. Protoc. 2013, 8, 148–161.
  70. Martinez Molina, D.; Jafari, R.; Ignatushchenko, M.; Seki, T.; Larsson, E.A.; Dan, C.; Sreekumar, L.; Cao, Y.; Nordlund, P. Monitoring drug target engagement in cells and tissues using the cellular thermal shift assay. Science 2013, 341, 84–87.
  71. Savitski, M.M.; Reinhard, F.B.; Franken, H.; Werner, T.; Savitski, M.F.; Eberhard, D.; Martinez Molina, D.; Jafari, R.; Dovega, R.B.; Klaeger, S.; et al. Tracking cancer drugs in living cells by thermal profiling of the proteome. Science 2014, 346, 1255784.
  72. Gaetani, M.; Sabatier, P.; Saei, A.A.; Beusch, C.M.; Yang, Z.; Lundstrom, S.L.; Zubarev, R.A. Proteome Integral Solubility Alteration: A High-Throughput Proteomics Assay for Target Deconvolution. J. Proteome Res. 2019, 18, 4027–4037.
  73. Kaur, S.; McGuire, L.; Tang, D.; Dollinger, G.; Huebner, V. Affinity selection and mass spectrometry-based strategies to identify lead compounds in combinatorial libraries. J. Protein Chem. 1997, 16, 505–511.
  74. Jonker, N.; Kool, J.; Irth, H.; Niessen, W.M. Recent developments in protein-ligand affinity mass spectrometry. Anal. Bioanal. Chem. 2011, 399, 2669–2681.
  75. Maecker, H.T.; McCoy, J.P.; Nussenblatt, R. Standardizing immunophenotyping for the Human Immunology Project. Nat. Rev. Immunol. 2012, 12, 191–200.
  76. Gratama, J.W.; Sutherland, D.R.; Keeney, M.; Papa, S. Flow cytometric enumeration and immunophenotyping of hematopoietic stem and progenitor cells. J. Biol. Regul. Homeost. Agents 2001, 15, 14–22.
  77. Kim, K.M.; Noh, J.H.; Bodogai, M.; Martindale, J.L.; Yang, X.; Indig, F.E.; Basu, S.K.; Ohnuma, K.; Morimoto, C.; Johnson, P.F.; et al. Identification of senescent cell surface targetable protein DPP4. Genes Dev. 2017, 31, 1529–1534.
  78. Herman, A.B.; Tsitsipatis, D.; Anerillas, C.; Mazan-Mamczarz, K.; Carr, A.E.; Gregg, J.M.; Wang, M.; Zhang, J.; Michel, M.; Henry-Smith, C.; et al. DPP4 inhibition impairs senohemostasis to improve plaque stability in atherosclerotic mice. J. Clin. Investig. 2023, 133, e165933.
  79. Amor, C.; Feucht, J.; Leibold, J.; Ho, Y.J.; Zhu, C.; Alonso-Curbelo, D.; Mansilla-Soto, J.; Boyer, J.A.; Li, X.; Giavridis, T.; et al. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 2020, 583, 127–132.
  80. Althubiti, M.; Lezina, L.; Carrera, S.; Jukes-Jones, R.; Giblett, S.M.; Antonov, A.; Barlev, N.; Saldanha, G.S.; Pritchard, C.A.; Cain, K.; et al. Characterization of novel markers of senescence and their prognostic potential in cancer. Cell Death Dis. 2014, 5, e1528.
  81. Poblocka, M.; Bassey, A.L.; Smith, V.M.; Falcicchio, M.; Manso, A.S.; Althubiti, M.; Sheng, X.; Kyle, A.; Barber, R.; Frigerio, M.; et al. Targeted clearance of senescent cells using an antibody-drug conjugate against a specific membrane marker. Sci. Rep. 2021, 11, 20358.
  82. Qu, A.; Wu, X.; Li, S.; Sun, M.; Xu, L.; Kuang, H.; Xu, C. An NIR-Responsive DNA-Mediated Nanotetrahedron Enhances the Clearance of Senescent Cells. Adv. Mater. 2020, 32, 2000184.
  83. Hoare, M.; Ito, Y.; Kang, T.W.; Weekes, M.P.; Matheson, N.J.; Patten, D.A.; Shetty, S.; Parry, A.J.; Menon, S.; Salama, R.; et al. NOTCH1 mediates a switch between two distinct secretomes during senescence. Nat. Cell Biol. 2016, 18, 979–992.
  84. Kim, K.M.; Noh, J.H.; Bodogai, M.; Martindale, J.L.; Pandey, P.R.; Yang, X.; Biragyn, A.; Abdelmohsen, K.; Gorospe, M. SCAMP4 enhances the senescent cell secretome. Genes Dev. 2018, 32, 909–914.
  85. Doolittle, M.L.; Saul, D.; Kaur, J.; Rowsey, J.L.; Vos, S.J.; Pavelko, K.D.; Farr, J.N.; Monroe, D.G.; Khosla, S. Multiparametric senescent cell phenotyping reveals targets of senolytic therapy in the aged murine skeleton. Nat. Commun. 2023, 14, 4587.
  86. Pauwels, J.; Fijalkowska, D.; Eyckerman, S.; Gevaert, K. Mass spectrometry and the cellular surfaceome. Mass Spectrom. Rev. 2022, 41, 804–841.
  87. Meyfour, A.; Pahlavan, S.; Mirzaei, M.; Krijgsveld, J.; Baharvand, H.; Salekdeh, G.H. The quest of cell surface markers for stem cell therapy. Cell. Mol. Life Sci. 2021, 78, 469–495.
  88. Wollscheid, B.; Bausch-Fluck, D.; Henderson, C.; O’Brien, R.; Bibel, M.; Schiess, R.; Aebersold, R.; Watts, J.D. Mass-spectrometric identification and relative quantification of N-linked cell surface glycoproteins. Nat. Biotechnol. 2009, 27, 378–386.
  89. Bausch-Fluck, D.; Hofmann, A.; Wollscheid, B. Cell surface capturing technologies for the surfaceome discovery of hepatocytes. Methods Mol. Biol. 2012, 909, 1–16.
  90. Berg Luecke, L.; Gundry, R.L. Assessment of Streptavidin Bead Binding Capacity to Improve Quality of Streptavidin-based Enrichment Studies. J. Proteome Res. 2021, 20, 1153–1164.
  91. van Oostrum, M.; Muller, M.; Klein, F.; Bruderer, R.; Zhang, H.; Pedrioli, P.G.A.; Reiter, L.; Tsapogas, P.; Rolink, A.; Wollscheid, B. Classification of mouse B cell types using surfaceome proteotype maps. Nat. Commun. 2019, 10, 5734.
  92. van Oostrum, M.; Campbell, B.; Seng, C.; Muller, M.; Tom Dieck, S.; Hammer, J.; Pedrioli, P.G.A.; Foldy, C.; Tyagarajan, S.K.; Wollscheid, B. Surfaceome dynamics reveal proteostasis-independent reorganization of neuronal surface proteins during development and synaptic plasticity. Nat. Commun. 2020, 11, 4990.
  93. Luecke, L.B.; Waas, M.; Littrell, J.; Wojtkiewicz, M.; Castro, C.; Burkovetskaya, M.; Schuette, E.N.; Buchberger, A.R.; Churko, J.M.; Chalise, U.; et al. Surfaceome mapping of primary human heart cells with CellSurfer uncovers cardiomyocyte surface protein LSMEM2 and proteome dynamics in failing hearts. Nat. Cardiovasc. Res. 2023, 2, 76–95.
  94. Muller, M.; Grabnitz, F.; Barandun, N.; Shen, Y.; Wendt, F.; Steiner, S.N.; Severin, Y.; Vetterli, S.U.; Mondal, M.; Prudent, J.R.; et al. Light-mediated discovery of surfaceome nanoscale organization and intercellular receptor interaction networks. Nat. Commun. 2021, 12, 7036.
  95. Waas, M.; Littrell, J.; Gundry, R.L. CIRFESS: An Interactive Resource for Querying the Set of Theoretically Detectable Peptides for Cell Surface and Extracellular Enrichment Proteomic Studies. J. Am. Soc. Mass Spectrom. 2020, 31, 1389–1397.
  96. Waas, M.; Snarrenberg, S.T.; Littrell, J.; Jones Lipinski, R.A.; Hansen, P.A.; Corbett, J.A.; Gundry, R.L. SurfaceGenie: A web-based application for prioritizing cell-type-specific marker candidates. Bioinformatics 2020, 36, 1367–4811.
  97. Omasits, U.; Ahrens, C.H.; Müller, S.; Wollscheid, B. Protter: Interactive protein feature visualization and integration with experimental proteomic data. Bioinformatics 2014, 30, 884–886.
  98. Bausch-Fluck, D.; Hofmann, A.; Bock, T.; Frei, A.P.; Cerciello, F.; Jacobs, A.; Moest, H.; Omasits, U.; Gundry, R.L.; Yoon, C.; et al. A Mass Spectrometric-Derived Cell Surface Protein Atlas. PLoS ONE 2015, 10, e0121314.
  99. Bausch-Fluck, D.; Goldmann, U.; Müller, S.; van Oostrum, M.; Müller, M.; Schubert Olga, T.; Wollscheid, B. The in silico human surfaceome. Proc. Natl. Acad. Sci. USA 2018, 115, E10988–E10997.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations