Protein phosphorylation on serine and threonine residues is a widely distributed post-translational modification on proteins that acts to regulate their function. Phosphoprotein phosphatases (PPPs) contribute significantly to a plethora of cellular functions through the accurate dephosphorylation of phosphorylated residues. Most PPPs accomplish their purpose through the formation of complex holoenzymes composed of a catalytic subunit with various regulatory subunits. PPP holoenzymes then bind and dephosphorylate substrates in a highly specific manner. Despite the high prevalence of PPPs and their important role for cellular function, their mechanisms of action in the cell are still not well understood. Nevertheless, substantial experimental advancements in (phospho-)proteomics, structural and computational biology have contributed significantly to a better understanding of PPP biology in recent years. This Review focuses on recent approaches and provides an overview of substantial new insights into the complex mechanism of PPP holoenzyme regulation and substrate selectivity.

Post-translational modifications (PTMs) are covalent, mostly reversible, modifications of amino acid residues in proteins (Khoury et al., 2011; Ramazi and Zahiri, 2021). To date, more than 400 different PTMs have been identified, with the majority being applied and removed by enzymes (Khoury et al., 2011; Ramazi and Zahiri, 2021). Reversible protein phosphorylation is a binary PTM that is substantially used for signal transduction (Hunter, 1995). This modification has an effect on virtually all biological functions and changes many properties of proteins, such as their stability, binding, structure, localization and activity (Brautigan and Shenolikar, 2018). It is mediated by the counterplay of protein kinases and protein phosphatases, and can be a highly dynamic process, as demonstrated by some phosphorylated sites (p-sites) having half-lives in the order of seconds (Kleiman et al., 2011; Blazek et al., 2015; Reddy et al., 2016). To understand the nature of reversible phosphorylation, it is important to keep in mind that p-sites are not continuously modified through the kinase–phosphatase interplay. The state of a p-site depends on its required function for the cell, and can be under fairly stable kinase or phosphatase dominance (Nasa and Kettenbach, 2018; Nilsson, 2019; Kauko et al., 2020; Swartz et al., 2021). Owing to the ubiquity and dynamic nature of protein phosphorylation, this type of PTM is well suited for modulating numerous cellular processes, including transcription, DNA damage response, RNA processing, nutrient and stress responses, immunity, growth and many more (Ubersax and Ferrell, 2007; Guccione and Richard, 2019; Yang et al., 2017; Nasa et al., 2020). Consequently, dysregulated (de-)phosphorylation is a hallmark of many diseases including cancer (Zanivan et al., 2013; Vainonen et al., 2021), Alzheimer's disease (Grundke-Iqbal et al., 1986; Eidenmüller et al., 2001; Oliveira et al., 2017), cardiovascular diseases (Nicolaou and Kranias, 2009; Fischer et al., 2018), and diabetes (Meyerovitch et al., 1989; Danielsson et al., 2005). Phosphoserine (pSer) and phosphothreonine (pThr) are among the most frequently identified PTMs (Khoury et al., 2011; Ramazi and Zahiri, 2021). The pSer/pThr-specific phosphoprotein phosphatases (PPPs) dephosphorylate the majority of Ser/Thr p-sites (Li et al., 2013; Damle and Köhn, 2019). Although some of the responsible kinases are understood well enough to serve as drug targets, an understanding of the counteracting PPPs still lags behind. Nevertheless, phosphatases are by now also considered druggable targets, as recently reviewed elsewhere (Köhn, 2020; Zhang et al., 2021). The cellular and physiological functions of individual PPPs have extensively been discussed in recent reviews (Ohama, 2019; Park and Lee, 2020; Nilsson, 2019; Sager et al., 2020; Sandal et al., 2021). In this Review, we summarize recent findings regarding the mechanism of specificity, newly developed experimental approaches and phosphoproteomic insights that contribute to a better understanding of PPPs. In terms of modulator development, a recent comprehensive review summarizes small-molecule approaches in the context of cancer (Zhang et al., 2021). In addition, different reviews have covered therapeutic targeting of phosphatases (Vainonen et al., 2021; Köhn, 2020). Therefore, here, we focus on the development of non-small molecule modulators, mention new developments for the previously reviewed small-molecule modulators, and highlight the most recent developments in research tool development. As PP3 (also known as calcineurin or PP2B) has extensively been reviewed elsewhere (Creamer, 2020; Chaklader and Rothermel, 2021), it is not included here. Furthermore, we also do not consider PP7 (encoded by PPEF1 and PPEF2) because limited progress in the areas covered here has been made since a previous review was published (Brautigan and Shenolikar, 2018).

Phosphorylation of serine and threonine residues is catalyzed by more than 400 protein Ser/Thr-specific kinases (Manning et al., 2002; Li et al., 2013). The opposing dephosphorylation of the respective pSer/pThr sites is executed by protein Ser/Thr-specific phosphatases. Most of these phosphatases belong to one of three evolutionary unrelated superfamilies: (1) the phosphoprotein phosphatases-like (PPPL) family, (2) the metal-dependent protein phosphatases (PPMs), or (3) the haloacid dehalogenase (HAD) phosphatases (Damle and Köhn, 2019). More than 90% of all pSer/pThr-sites are thought to be dephosphorylated by the PPP family, a subfamily of the PPPLs, which establishes them as the major counteractors of pSer/pThr phosphorylation in eukaryotes (Brautigan and Shenolikar, 2018). This ability to regulate the vast majority of the highly complex pSer/pThr network in the cell is all the more fascinating given that this family consists only of seven phosphatases, namely PP1 (encoded by the three isoforms PPP1CA, PPP1CB and PPP1CC), PP2A (PPP2CA and PPP2CB), PP3 (PPP3CA, PPP3CB and PPP3CC), PP4 (PPP4C), PP5 (PPP5C), PP6 (PPP6C) and PP7 (PPEF1 and PPEF2). (Damle and Köhn, 2019).

Superficially, there is broad similarity between the different PPPs. The core catalytic protein is well conserved between PPPs, and there is a high overall degree of sequence identity across all eukaryotic species (Brautigan and Shenolikar, 2018). Inside their active sites, most PPPs coordinate an Fe2+ and a Zn2+ ion as essential cofactors through three highly conserved sequence stretches (-GDXHG-, -GDXVDRG- and -GNHE-) (Nasa and Kettenbach, 2018). Furthermore, most of the earliest discovered chemical inhibitors bind the catalytically active sites of several PPPs and act as pan-inhibitors with nanomolar to micromolar affinities (Zhang et al., 2021, 2013). Nevertheless, PPPs differ in the formation of their numerous holoenzymes, which are the functional entities in cells and consist of catalytic (denominated ‘c’ compared to the general holoenzyme, e.g. ‘PP1c’ and ‘PP1’) and a great variety of noncatalytic subunits (examples in Fig. 1A,B) (Brautigan, 2013; Li et al., 2013). PP5 is an exception as it is a multidomain enzyme (Fig. 1C). Regulatory subunits dictate holoenzyme activity by determining its subcellular localization, activity, binding partners, and to a large extent their substrate specificity. PPPs that act as free catalytic subunits, rather than holoenzymes within the cell, have not been reported. The assembly of catalytic subunits and PPP-specific regulatory subunits results in a highly specific biomolecular machinery, where each holoenzyme constitutes an independent signaling entity. This not only raises the complexity of PPP signaling networks and therefore complicates detailed studies (Needham et al., 2019; Fahs et al., 2016), but also leads to a similar number of distinct PPP complexes and Ser/Thr-specific kinases in eukaryotic cells. Despite the importance of holoenzyme formation by regulatory subunits, few mechanisms of PPP control by these regulatory subunits have been investigated in detail.

Fig. 1.

PP1 and PP2A exist as holoenzymes, whereas PP5 is a multidomain protein. Catalytic site ions are illustrated as red spheres within the catalytic domains (gray) of the represented PPP structures. Catalytic subunits/domain are slightly transparent to visualize the ion pair. For orientation, the active side is marked in red when metal ions are not resolved. (A) PP1 forms mostly dimeric or trimeric holoenzymes with diverse regulatory subunits (PDB IDs from left to right: 6ZEJ, 7UPI, 1S70, 7NZM and 2O8A) leaving the active site open (MYPT1, Phactr1, SHOC2 and PPP1R15A) or closed (Inhibitor-2). (B) PP2A forms trimeric holoenzymes. The catalytic subunit (gray) and the scaffolding A-subunit (orange) form an invariant core enzyme. The B-subunits [here B56 (purple) or B55 (light purple)] bind to the core enzyme and serve as regulatory subunits (PDB IDs from left to right: 2NPP and 3DW8). (C) PP5 does not form holoenzymes but exists as a multidomain protein. The N-terminal TPR domains (dark gray) of PP5 shield the ion pair at the catalytic site, leaving it sterically inaccessible to substrates (PDB ID: 1WAO). This conformation opens up upon activation of PP5.

Fig. 1.

PP1 and PP2A exist as holoenzymes, whereas PP5 is a multidomain protein. Catalytic site ions are illustrated as red spheres within the catalytic domains (gray) of the represented PPP structures. Catalytic subunits/domain are slightly transparent to visualize the ion pair. For orientation, the active side is marked in red when metal ions are not resolved. (A) PP1 forms mostly dimeric or trimeric holoenzymes with diverse regulatory subunits (PDB IDs from left to right: 6ZEJ, 7UPI, 1S70, 7NZM and 2O8A) leaving the active site open (MYPT1, Phactr1, SHOC2 and PPP1R15A) or closed (Inhibitor-2). (B) PP2A forms trimeric holoenzymes. The catalytic subunit (gray) and the scaffolding A-subunit (orange) form an invariant core enzyme. The B-subunits [here B56 (purple) or B55 (light purple)] bind to the core enzyme and serve as regulatory subunits (PDB IDs from left to right: 2NPP and 3DW8). (C) PP5 does not form holoenzymes but exists as a multidomain protein. The N-terminal TPR domains (dark gray) of PP5 shield the ion pair at the catalytic site, leaving it sterically inaccessible to substrates (PDB ID: 1WAO). This conformation opens up upon activation of PP5.

Technical advancements in recent years have helped establish global high-throughput approaches to better decipher PPP biology. Structural insights coupled with phospho-proteomics, or chemical molecule screens followed by computational analysis have provided novel insights. Emerging insights comprise the validation of intrinsic substrate specificity of catalytic subunits, the short linear motif (SLiM)-based interaction between catalytic and non-catalytic subunits, as well as between the holoenzymes and their interactors (Heroes et al., 2013; Hertz et al., 2016; Ueki et al., 2021; Fowle et al., 2021). The SLIM-mediated interaction is a universal mechanism through which phosphatases and kinases recognize and bind to their interactors as well as substrates (Seok, 2021). In addition, a new chemical proteomics approach that uses active site inhibitor–bait pulldowns followed by mass spectrometry (PIB-MS) read-out enables the specific identification of the PPP interactome in different conditions (Lyons et al., 2018; Nasa et al., 2020). Furthermore, new discoveries and developments of inhibitors or activators for PPPs now make it possible to distinguish certain PPPs in cellular experiments and target them specifically over other PPP members. Recent findings on interactions, mechanisms and methodologies for studying each of the different PPPs are discussed in the following sections.

Attributed to the advances of global approaches in mass spectrometry (MS), the number of identified p-sites of the human phosphoproteome has expanded exponentially over the past two decades. This has enabled scientists to uncover the extraordinary scale and complexity of the phosphoproteome (Khoury et al., 2011; Needham et al., 2019; Ochoa et al., 2019; Ramazi and Zahiri, 2021). These studies have revealed that most proteins are phosphorylated, many on multiple sites (Olsen et al., 2010; Sharma et al., 2014; Needham et al., 2019). Ochoa and colleagues generated the most complete human phosphoproteome synopsis to date. Their analysis of 112 human phosphoproteome datasets found 11,982 proteins bearing 119,809 phosphosites (Ochoa et al., 2019). Analysis of the residue-specific phosphorylation distribution through the dbPTM database unveiled that pTyr makes up only a smaller proportion (∼11%) of the total phosphorylations, whereas most of the phosphorylations are found on pThr (∼22%) and pSer (∼67%) residues (Ramazi and Zahiri, 2021). Phosphorylations on other residues exist but are scarce (Ramazi and Zahiri, 2021). This plethora of Ser and Thr phosphorylations highlights the importance of spatio-temporal regulation of PPPs for these modifications (Cohen, 2002; Khoury et al., 2011; Nilsson, 2019).

Although phosphorylation has been repeatedly found to be essential for proper function of the cell, more than 97% of identified human p-sites have no assigned function (Needham et al., 2019; Ramazi and Zahiri, 2021). Usually, substantial effort, such as thorough low-throughput experiments, is needed to uncover the individual p-site function. Thus, although we can ‘quantify’ the phosphoproteome, we are still far from understanding it. One approach to rectifying this involves classifying p-sites according to their position in an ordered region (OR) or intrinsically disordered region (IDR) of a protein. However, the p-sites of IDRs are even less characterized than the ordered sites based on follow-up experiments, because their intrinsic flexibility results in less structural information. Recent results show that ∼40% of the eukaryotic proteome consists of IDRs, and that these segments contain phosphorylation signaling hubs (Wright and Dyson, 2015), suggesting that conformational heterogeneity might be important to kinase as well as phosphatase selectivity. Furthermore, it has been observed that the phosphorylation state of IDRs correlates positively with IDR length (Fukuchi et al., 2011; Cho et al., 2020; Koike et al., 2020). Interestingly, initial studies have suggested that p-sites within ORs more often regulate enzyme activity and conformation, whereas IDRs more often contain p-sites that regulate the localization of a protein (Ochoa et al., 2019; Zarin et al., 2021).

Great advancements in recent years have led to the development of computational methods that have helped to predict ORs and IDRs within protein structures (Jumper et al., 2021; Chakravarty and Porter, 2022) and gained a deeper readout of the human phosphoproteome. However, most functions of the p-sites remain to be elucidated, and predicting and assigning them is still challenging (Kokot et al., 2022; Chaudhari et al., 2021). To change this, a detailed understanding of PPP biology and the development of global PPP approaches are essential. The first recently established global approaches to determine p-site assignment, regulation and function are listed in Table 1.

Table 1.

Large-scale phosphoproteomic studies conducted with PPPs or their interaction partner

Large-scale phosphoproteomic studies conducted with PPPs or their interaction partner
Large-scale phosphoproteomic studies conducted with PPPs or their interaction partner

PP1 is ubiquitously expressed and is considered one of the major PPPs because it counteracts more than 100 kinases (Li et al., 2013). The catalytic subunit (PP1c) forms transient holoenzymes with one or two regulatory proteins to fulfill this comprehensive role. These regulatory proteins (>200) are known as regulatory interactors of protein phosphatase one (RIPPOs) (Bollen et al., 2010; Wu et al., 2018). The formation of these transient holoenzymes coordinates the spatiotemporal activity of PP1 and its substrate specificity (Bollen et al., 2010). Although the regulatory function of RIPPOs is undisputed, it has been speculated that PP1c itself contributes to substrate specificity of PP1 (Ragusa et al., 2010; Rogers et al., 2016). PP1c has three substrate-binding grooves – one acidic, one hydrophobic and one C-terminal groove that radiate in a Y-shaped pattern from the active site and supposedly interact with substrates for dephosphorylation (Fig. 2A) (Peti and Page, 2015; Ragusa et al., 2010; Choy et al., 2014; Peti et al., 2013). Notably, when bound to substrate-targeting RIPPOs, the active site of PP1c remains largely accessible for the substrate (Fig. 1A) (Fedoryshchak et al., 2020; Chen et al., 2015; Kwon et al., 2022). This is in contrast to PP2A–B-subunit holoenzymes where the B-subunit is in direct proximity to the active site (Xu et al., 2006, 2008). A recent crystal structure of the Phactr1–PP1c holophosphatase shows a remodeling of the hydrophobic groove, which is engaged by the short PP1 substrate peptide from the insulin receptor tyrosine kinase substrate of 53 kDa (IRSp53; also known as BAIAP2) (Fig. 2D), leaving the acidic groove available for electrostatic interactions (Fedoryshchak et al., 2020). More broadly, we recently demonstrated the intrinsic substrate specificity of PP1c by combining a phosphopeptide library dephosphorylation followed by mass spectrometry approach (PLDMS) with phosphoproteomics, which offered a range of substrate candidates that can be dephosphorylated by PP1c (Hoermann and Köhn, 2021; Hoermann et al., 2020). We validated an earlier observed preference of basophilic residues N-terminal of the p-site (Lee et al., 1999; Li et al., 2013) and reported a faster dephosphorylation of pThr compared to pSer (Hoermann et al., 2020). Our follow-up analysis demonstrated that the preference of the basophilic sequence context surrounding the p-site is enhanced when located within IDRs, which lack features for structural recognition (Fig. 2B) (Hoermann and Köhn, 2021). Thus, in IDRs, the amino acid sequence context gains importance for site targeting. Interestingly, in the case of the structured PP1 substrate translation initiation factor 2α (eIF2α), which was resolved by CryoEM together with the PP1c–PPP1R15A–G-actin complex, the substrate recruitment is mediated by PPP1R15A–G-actin (Fig. 2D), and PPP1R15A occupies the C-terminal groove of PP1c, whereas eIF2α comes close to the active pocket and does not occupy the grooves (Yan et al., 2021). Taken together, these results suggest an intrinsic catalytic subunit-derived specificity of p-sites within IDRs, whereas recognition of sites in ORs might be more RIPPO driven.

Fig. 2.

RIPPO-binding sites and dephosphorylation site preference of PP1. (A) The three substrate-binding grooves of PP1c radiate outward from its active site. PP1c binds its substrates through a combination of two of these binding grooves. Holoenzyme formation can influence the shape of the grooves or block them, but there are no examples yet for acidic groove blockage. Metal ions illustrated as red spheres (PDB ID: 4XPN). (B) PP1c-sensitive sites of ordered and disordered regions are characterized by the positively charged amino acids arginine and lysine found N-terminally to the phosphorylation site. Panel adapted from Hoermann and Köhn (2021), where it was published under a CC-BY 4.0 license. (C) Binding sites on the surface of PP1c (PDB ID: 4XPN) that interact with RIPPOs are colored by residue. Selected binding site interactors are annotated according to their effect on PP1. Bold and underlining denote increased and inhibited activity, respectively; non-proteinaceous interacting partners are italicized. Most RIPPOs interact through a combination of more than one motif. (D) Structures of PP1 holoenzymes with substrates bound to the active site of PP1c. Phactr1 remodels the hydrophobic groove of PP1c for better IRSp53 peptide binding (PDB ID: 6ZEJ). PPP1R15A brings PP1c and G-actin together in order to coordinate dephosphorylation of the N-terminal domain or phosphorylated eIF2α (eIF2αP-NTD; PDB ID: 7NZM).

Fig. 2.

RIPPO-binding sites and dephosphorylation site preference of PP1. (A) The three substrate-binding grooves of PP1c radiate outward from its active site. PP1c binds its substrates through a combination of two of these binding grooves. Holoenzyme formation can influence the shape of the grooves or block them, but there are no examples yet for acidic groove blockage. Metal ions illustrated as red spheres (PDB ID: 4XPN). (B) PP1c-sensitive sites of ordered and disordered regions are characterized by the positively charged amino acids arginine and lysine found N-terminally to the phosphorylation site. Panel adapted from Hoermann and Köhn (2021), where it was published under a CC-BY 4.0 license. (C) Binding sites on the surface of PP1c (PDB ID: 4XPN) that interact with RIPPOs are colored by residue. Selected binding site interactors are annotated according to their effect on PP1. Bold and underlining denote increased and inhibited activity, respectively; non-proteinaceous interacting partners are italicized. Most RIPPOs interact through a combination of more than one motif. (D) Structures of PP1 holoenzymes with substrates bound to the active site of PP1c. Phactr1 remodels the hydrophobic groove of PP1c for better IRSp53 peptide binding (PDB ID: 6ZEJ). PPP1R15A brings PP1c and G-actin together in order to coordinate dephosphorylation of the N-terminal domain or phosphorylated eIF2α (eIF2αP-NTD; PDB ID: 7NZM).

Addressing the challenge of assigning a specific holoenzyme to a substrate, Wu et al. developed a PP1c-holoenzyme-fusion based substrate trapping approach with reduced PP1 activity (Wu et al., 2018). Together with the substrate candidates from the phosphoproteomics approach (Hoermann et al., 2020), these developments offer novel possibilities to decipher the PP1 holoenzyme substratome.

Many interactions between PP1c and RIPPOs are governed by the surface features of PP1c, which are distributed as specific sites almost over its entire surface and distinguish PP1c from other PPP-type phosphatases (Fig. 2C) (Verbinnen et al., 2017; Bollen et al., 2010; Brautigan and Shenolikar, 2018). It is understood that most RIPPOs use intrinsically disordered PP1-binding domains with one or more SLiMs to dock to these particular sites on the surface of PP1c. The most common PP1-binding SLiMs are variants of the RVxF, SILK, myosine phosphatase N-terminal element (MyPhoNE) and ϕϕ (two consecutive hydrophobic residues) motifs (Terrak et al., 2004; Peti and Page, 2015; Bollen et al., 2010). A remarkable amount of RIPPOs (∼150) carry the RVxF motif (Meiselbach et al., 2006), which binds to two hydrophobic pockets on the opposite side to the active site of PP1c (Wakula et al., 2003; Egloff et al., 1997). The PP1-specific RVxF binding motif was ideal for developing chemical modulators of PP1, enabling the creation of selective PP1 modulators, so called PP1-disrupting peptides (PDPs; see Box 1) (Chatterjee et al., 2012). So far, no cleanly PP1-specific chemical inhibitor has been reported, as all also inhibit other PPPs at the higher concentrations that are commonly used in cellular applications. However, the small-molecule inhibitor Tautomycetin is fairly specific (Choy et al., 2017), and the overexpression of endogenous inhibitors of PP1, such as nuclear inhibitor of PP1 (NIPP1; also known as PPP1R8) or inhibitor-1–3 (I1, I2 and I3; also known as PPP1R1, PPP1R2 and PPP1R11) (Fig. 1A), which use SLiMs in PP1 as well, are often used to inhibit the catalytic subunit of PP1 over other PPPs in cellular experiments (Krause et al., 2018; Winkler et al., 2015).

Box 1. New research tools for PPPs

The assembly of regulatory interactors of protein phosphatase one (RIPPOs) with PP1 is commonly mediated by specific short linear motifs (SLiMs) on the surface of the RIPPOs. The so-called RVxF motif (x is any amino acid except proline) is the most widely used motif involved in the PP1–RIPPO interaction (Bollen et al., 2010). The ∼20-mer PP1-disrupting peptides (PDPs) bind the respective pocket on PP1, thereby releasing it from a subset of RIPPOs, and enabling the dephosphorylation of nearby substrates directed by the intrinsic PP1c substrate specificity (Chatterjee et al., 2012; Hoermann et al., 2020). So far, PDPs are the only selective chemical modulators of PP1c, as they do not bind to other PPPs. They have been used to study PP1 substrates in the MAPK pathway (Wang et al., 2019b), the role of PP1 role in Ca2+ release (Reither et al., 2013) and for activation of PP1 to treat arrhythmia and heart failure (Fischer et al., 2018). Addressing the need for a more controlled modulation of PP1c activity, a photo-releasable version named PDP-caged was developed, which can be light-controlled in living cells (Trebacz et al., 2020).

In another approach, PDP1 was coupled to an AKT inhibitor to form a heterobifunctional phosphatase-recruiting chimera (PhoRC), recruiting PP1 to AKT and inducing its dephosphorylation (Yamazoe et al., 2020).

The concept of phosphorylation-targeting chimeras (PhosTACs) is similar to PhoRCs and based on proteolysis-targeting chimeras (PROTACs) (Sakamoto et al., 2001; Chen et al., 2021). Like ProTACs, PhosTACs are heterobifunctional small molecules that induce complex formation and are composed of two protein-binding regions connected through a linker. In contrast to ProTACs, PhosTACs do not induce target degradation. Instead, PhosTACs, like PhoRCs, induce proximity-dependent dephosphorylation of the substrate by mediating the interaction between the substrate and a phosphatase (see figure, which shows a cartoon-based representation of the PhosTAC approach). The selected Ser/Thr phosphatase is recruited by one part of the PhosTAC, while the opposing region captures the targeted substrate. Chen et al. generated a construct, in which the PP2A A-subunit is fused with FKBP12(F36V), and a Halotag7 (abbreviated as Halo) is fused to the Forkhead-box O3a (FOXO3a) transcription factor, a known PP2A substrate (Chen et al., 2021). They successfully demonstrated the dephosphorylation-dependent transcriptional activation of a reporter gene upon treatment with the bifunctional molecule containing a Rapalog [which binds to FKBP(F36V)] and a Halotag substrate (which binds to the Halotag), which induced FOXO3a dephosphorylation and activation (Chen et al., 2021). An advantage of PhosTACs is that they are not limited to inducing a loss-of-function effect of the targeted protein, as in the case for ProTACs, but can also trigger a gain-of-function effect depending on the nature of the targeted p-site. PhosTACs have great potential to determine the precise effect of p-sites by minimizing off-target effects. Additionally, they fulfill the need of an alternative strategy to the use of kinase inhibitors to identify targetable dysregulated phosphorylation activity in clinical applications.

Not all RIPPOs use SLiMs to bind to PP1. For example, to dephosphorylate RAF in the mitogen-activated protein kinase (MAPK) pathway, SHOC2 binds PP1c and MRAS through the concave surface of its leucine-rich repeat region, and only contains a cryptic RVxF motif (Fig. 1A) (Kwon et al., 2022; Hauseman et al., 2022). Similarly, the suppressor-of-Dis2-number 2 (SDS22; also known as PPP1R7) has no known SLiM and associates with PP1c through structured domains, such as helical leucine repeats, which interact with a broad surface of PP1 (Ceulemans et al., 2002; Heroes et al., 2019). SDS22 and I3 form a ternary complex with PP1, and are widely expressed and conserved RIPPOs (Cao et al., 2021; Lesage et al., 2007; Choy et al., 2019). They control multiple steps of the life cycle of PP1. Indeed, they contribute to (1) the stabilization and activation of newly translated PP1, (2) the translocation of PP1 to the nucleus, and (3) the storage of PP1 as a reserve for holoenzyme assembly (Cao et al., 2021). Preliminary evidence suggests that SDS22 and I3 might also function as scavengers of released or aged PP1 for re-use in holoenzyme assembly or proteolytic degradation, respectively (reviewed in Cao et al., 2021).

Phosphoprotein phosphatase 2A (PP2A) is the second major PPP alongside PP1. It is ubiquitously expressed and contributes up to 1% of the total cellular protein in mammalian cells (Ruediger et al., 1991). Canonically, it acts as a heterotrimeric holoenzyme consisting of the catalytic (PP2Ac) and a scaffolding (A) subunit, which form the dimeric, relatively invariant core enzyme. The core enzyme assembles with an exchangeable regulatory (B) subunit for functional phosphatase activity. So far, two multimeric non-canonical PP2A holoenzymes named STRIPAK and INTAC are described (Kück et al., 2019; Jeong et al., 2021; Zheng et al., 2020). Surprisingly, in the INTAC holoenzyme, Integrator8 (Int8; also known as INTS8) binds to the A-subunit in a B-subunit typical manner (Zheng et al., 2020). This is the first case of a PP2A holoenzyme without one of the established regulatory proteins, and the potential of integrators as a new regulatory subunit family need to be further investigated. The assembly of the variable and interchangeable B-subunit to the core enzyme results in a multitude of distinct PP2A holoenzymes (Cho and Xu, 2007). The B-subunits are subdivided into four major families – B, B′, B″ and B‴, with each comprising two to five isoforms (Fig. 3B) (Virshup and Shenolikar, 2009; Wlodarchak and Xing, 2016). They are key determinants of PP2A-substrate specificity due to their interaction with substrates. In contrast to the A- and C-subunit, isoforms of the distinct B-subunits vary strongly in their structure and function (Baharians and Schönthal, 1998; Xu et al., 2006; Slupe et al., 2011). Additionally, their expression levels alter throughout development, tissue and according to cellular requirement, whereas the dimeric core enzyme tends to be consistently and ubiquitously expressed (Ruediger et al., 1991; Seshacharyulu et al., 2013). In contrast to PP1, the structure of the PP2A holoenzyme is predominantly determined by the A and B-subunits, leaving less surface area of PP2Ac available for other interactions (Figs 1, 3A).

Fig. 3.

PP2A modulator target sites and different site preference upon B-subunit binding. (A) Residues on the surface of PP2Ac that interact with its holoenzyme subunits and other modulators are colored (PDB ID: 2NPP). Binding partners that increase PP2A activity are shown in bold and those that inhibit PP2A are underlined; non-proteinaceous partners are italicized. (B) Overview of the regulatory subunits of PP2A that bind to the core dimer. So far one interaction of an integrator as a regulatory subunit has been reported, which adds a new subunit family to the regulators of PP2A. (C) IceLogos visualizing the conserved amino acid patterns of sites dephosphorylated by PP2A–B55 and PP2A–B56 holoenzymes after incubation with cell lysate. The sequence context of dephosphorylated sites is determined though the B-subunit. Adapted from Kruse et al. (2020), where it was published under a CC-BY 4.0 license.

Fig. 3.

PP2A modulator target sites and different site preference upon B-subunit binding. (A) Residues on the surface of PP2Ac that interact with its holoenzyme subunits and other modulators are colored (PDB ID: 2NPP). Binding partners that increase PP2A activity are shown in bold and those that inhibit PP2A are underlined; non-proteinaceous partners are italicized. (B) Overview of the regulatory subunits of PP2A that bind to the core dimer. So far one interaction of an integrator as a regulatory subunit has been reported, which adds a new subunit family to the regulators of PP2A. (C) IceLogos visualizing the conserved amino acid patterns of sites dephosphorylated by PP2A–B55 and PP2A–B56 holoenzymes after incubation with cell lysate. The sequence context of dephosphorylated sites is determined though the B-subunit. Adapted from Kruse et al. (2020), where it was published under a CC-BY 4.0 license.

The limited number of assigned p-sites for a certain PP2A holoenzyme narrows our understanding of the mechanisms of holoenzyme-derived site-specific dephosphorylation. Recently, the first PP2A–B56 specific consensus binding motif, LxxIxE, was identified (Hertz et al., 2016). Structural analysis revealed that the LxxIxE SLiM engages with a well-conserved binding pocket, which is composed of a hydrophobic and a basic patch on opposite ends, and is present on all B56 isoforms (Hertz et al., 2016). Follow-up approaches exposed the requirement of a basic stretch adjacent to the LxxIxE motif on B56 interactors to enhance PP2A–B56 binding (Wang et al., 2020). After determining potential B56-binding peptide sequences using phage display libraries, more than 100 potential B56-binding partners were reported (Wu et al., 2017). Interestingly, Cip2A, a oncogenic inhibitor of PP2A, was found among them (Junttila et al., 2007; Laine et al., 2021). The idea of the LxxIxE motif as a PP2A–B56-specific inhibitor was successfully applied in a recent approach by overexpressing the SLiM as a peptide modulator (Kruse et al., 2020). Using this approach, the authors observed that PP2A–B56 assembly does not only serve substrate recruitment, but B56 binding to the catalytic subunit directly affects the sequence preference of the dephosphorylated p-site, revealing a selectivity for basophilic charged residues N-terminal of the p-site and evasion of proline-directed (P at +1) motifs (Fig. 3C; Kruse et al., 2020).

The avoidance of proline-directed motifs was also observed simultaneously in our study of the sole catalytic subunit (PP2Ac) at both the peptide and protein level (Hoermann et al., 2020). However, the basophilic selectivity of PP2A–B56 (Kruse et al., 2020) disagrees with the acidic-residue preference we noticed for PP2Ac (Hoermann et al., 2020). Furthermore, no selectivity of pThr over pSer was observed for PP2A–B56, which was detected for PP2Ac and the PP2A–B55 complex (Hoermann et al., 2020; Kruse et al., 2020). Interestingly, proline-directed motifs are targeted by PP2A–B55, but PP2A–B55 does not show a basophilic preference, resulting in significantly different p-sites being dephosphorylated by PP2A–B56 and PP2A–B55 (Fig. 3C; Kruse et al., 2020). These results show that the regulatory subunit of PP2A directly determines the p-site specificity of the holoenzyme, which results in unique patterns of kinase opposition (Kruse et al., 2020; Cundell et al., 2016; Jong et al., 2020). These findings go hand in hand with the observations in budding yeast from the Uhlmann laboratory (Godfrey et al., 2017; Touati et al., 2019). They observed that PP2A–Cdc55 (the PP2A–B55 homolog) counteracts proline-directed motifs and threonine phosphorylation while PP2A–Trs1 (the PP2A–B56 homolog) opposes basophilic p-site motifs. It is interesting to note that the respective subunits appear to structurally modify the active site and its selectivity, which raises the question as to what extent the mere catalytic subunit should be used for further selectivity experiments.

It has been known for some time that the acidic surface of PP2A–B55 binds to clusters of basic residues surrounding the phosphorylation site (Cundell et al., 2016). More recently, a binding motif for PP2A–B55 was identified on the basis of the substrate retinoblastoma-like protein 1 (p107, also known as RBL1) (Fowle et al., 2021; Jayadeva et al., 2010). Acidic residues of B55α (especially Asp197) bind to the (first arginine of the) conserved SLiM ‘[RK]Vxx[VI]R’, which is located four to 10 amino acids C-terminal of the proximal p-site (Kruse et al., 2020; Fowle et al., 2021). Interestingly, the identified sites bearing the motif validate the B55α-mediated preference of Ser/Thr-Pro-directed kinase motifs (Cundell et al., 2016; Zhao et al., 2019).

The assembly of B-subunits with the core enzyme is additionally regulated through PTMs on the unstructured C-terminal end of PP2Ac. About 70–90% of endogenous PP2A is C-terminally carboxymethylated (Lee and Pallas, 2007; Yu et al., 2001), which mediates the assembly of B55, B56 and B72 with the core dimer. Whereas the methylation appears to be crucial for B55 binding, B56 and B72 can also bind to unmethylated PP2Ac but prefer methylated forms (Yabe et al., 2018). Insufficient PP2A holoenzyme formation caused by the absence of carboxymethylation results in loss of its functionality as an important tumor suppressor (Fowle et al., 2019). Indeed, loss of carboxymethylation or removal of the C-terminal leucine residue can lead to elevated binding of striatin family B-subunits to the AC dimer, and this trimer can have oncogenic activities (Kim et al., 2020; Kurppa and Westermarck, 2020). Increased phosphorylation of Thr304 and Tyr307, as well as decreased methylation at these residues, appears to result in decreased activity of PP2A (Lee and Pallas, 2007; Janssens et al., 2008; Sangodkar et al., 2016; Löw et al., 2014). However, although the results of the C-terminus-mediated PP2A holoenzyme formation are consistent throughout the literature, the Ogris laboratory have shown that commercially available PP2A antibodies target the conserved C-terminus of the PP2A-like PPPs and do not exclusively bind to PP2Ac. Therefore, existing results need to be carefully assessed in light of the antibody issues (Frohner et al., 2020; Schüchner et al., 2020).

Owing to its prominent role in cancer (Ruvolo, 2019; Vainonen et al., 2021; Westermarck and Neel, 2020), PP2A is a popular target for small-molecule approaches (Zhang et al., 2021). One of these is the development of proximity-inducing molecules for targeted dephosphorylation (see Box 1). Moreover, small molecules have been postulated to selectively control the activity of PP2A by functioning as a molecular glue between the subunits (Morita et al., 2020; Leonard et al., 2020). However, new data reports on strong side effects and questions whether their effects can be interpreted as PP2A specific (Vit et al., 2022), resulting in previous findings now being reconsidered (Morita et al., 2022). Further studies are needed to evaluate these findings, but for now the biological conclusions drawn from utilizing these molecules should be considered in light of the available specificity data, and their use should be carefully planned in terms of conditions used.

Within the conserved PPP family, PP2A, PP4 and PP6 constitute the PP2A-like subfamily, as they are more closely related in sequence to one another than they are to the other PPPs (Andreeva and Kutuzov, 2001; Li et al., 2013). Despite their high sequence similarity, knockdown of the individual PP2A-like PPPs in C. elegans results in different phenotypes. This indicates that they fulfill separate functions despite structural similarities of the catalytic subunits (Kao et al., 2004; Han et al., 2009; Afshar et al., 2010; Martin-Granados et al., 2008). The catalytic subunit (PP4c) forms either a dimer with PPP4R1 or PPP4R4, or a trimer with PPP4R2 and PPP4R3α or PPP4Rβ (PPP4R3α/β) (Cohen et al., 2005; Gingras et al., 2005; Park and Lee, 2020). Currently, no three-dimensional structures of PP4 have been resolved, which hampers the identification of binding grooves or SLiMs as was achieved for PP1 and PP2A. Furthermore, compared to the two aforementioned major PPPs, little is known about the regulatory mechanisms of PP4 in the cell (Park and Lee, 2020). Nevertheless, PP4 has emerged as a nuclear and chromatin-associated phosphatase that is implicated in regulating numerous nuclear processes (Isono et al., 2017; Su et al., 2017; Youn et al., 2018; Wang et al., 2019a). Still, the nuclear import mechanism of PP4 remains to be elucidated, and it has not yet been determined which holoenzyme constellations of PP4 are imported into the nucleus.

Recently, binding partners of the trimeric PP4c–PPP4R2–PPP4R3α/β holoenzymes have been identified by using proteomic phage display (ProP-PD). The subunit PPP4R3 selectively recognizes binding partners containing FxxP and MxPP motifs (Lipinszki et al., 2015; Ueki et al., 2019). The binding pocket of these motifs is conserved in the N-terminal enabled/VASP homology 1 (EVH1) domain of PPP4R3. EVH1 domains are known to bind to proline-rich sequences (Ball et al., 2002). By determining the structure of the bound peptide, the phenylalanine and proline residues were demonstrated as the significant contact points of FxxP/MxPP motifs (Ueki et al., 2019). Owing to the abundance of FxxP sequences in the proteome, it is unlikely these will be the only determining factor in the PPP4R3–substrate interaction. It is expected that other as-yet-unidentified factors will contribute to this mechanism. PPP4R3 is furthermore the subunit that directly interacts with the PP4 inhibitor CT45, which reduces the activity of PP4 significantly but does not alter its interactome (Coscia et al., 2018).

Regarding a different aspect of substrate specificity, PP4c has been shown to dephosphorylate pThr faster than pSer in vitro (Ueki et al., 2019). This was also found to be the case for PP1c and PP2Ac, and concurs with the order of dephosphorylation observed in biological pathways (Hein et al., 2017; Hoermann et al., 2020). A notable exception is the above discussed PP2A:B56 holoenzyme (Kruse et al., 2020).

Post-translational modifications of PP4c and its regulatory subunits also have an impact on the activity of PP4 (Voss et al., 2013; Huang et al., 2016). Similar to the other PP2A-like phosphatases, the PP4c holoenzyme assembly is partially regulated through carboxymethylation of PP4c (Lyons et al., 2021). Of note, Lyons and colleagues nicely circumvent the above mentioned PP2A-antibody pitfalls through MS-based quantification of the PP2A-like phosphatases. To date, no PP4-specific chemical modulator has been reported, but the first endogenous inhibitors of PP4 were identified recently (Coscia et al., 2018; Han et al., 2015; Rosales et al., 2015; Park et al., 2019). Further studies of underlying mode of actions of these inhibitors might aid the development of PP4-specific chemical modulators.

Similar to other PPPs, PP5 is a ubiquitously expressed phosphatase important for a multitude of signaling pathways involved in cell cycle progression, stress responses, muscle contraction and other processes (Hinds and Sánchez, 2008; Sager et al., 2020; Krysiak et al., 2018; Neumann et al., 2021). In contrast to the other PPPs, PP5 is a multidomain phosphatase and does not form multimeric holoenzymes. The catalytic domain (PP5c) with its regulatory domain is encoded in a single gene throughout eukaryotes and forms one polypeptide (Chen et al., 2017). PP5c is N-terminally flanked by three tetratricopeptide repeat (TPR) domains, which bind to the C-terminal αJ-helix (Yang et al., 2005). Structural analysis revealed that together they maintain PP5 in an autoinhibited conformation with its active site covered by the TPR domains (Fig. 4A) (Yang et al., 2005). Interestingly, the interaction of the TPR helices with the PP5c domain resembles the mechanism of PP1c inhibition through pan-inhibitors (Yang et al., 2005; Kita et al., 2002; Maynes et al., 2001; Goldberg et al., 1995). Consequently, removal of the TPR domains or the C-terminal 13 residues potently activates the enzyme 10- to 50-fold (Yang et al., 2005; Oberoi et al., 2016). However, the TPR domain does not exclusively mediate the autoinhibition, but additionally is the primary interface for protein–protein interactions, substrate recruitment and phosphatase activation. Because of this versatility, the TPR domains of PP5 have emerged as the universal regulator of PP5.

Fig. 4.

TPR-domain-dependent activation of PP5. (A) Residues on the surface of PP5 (PDB ID: 1WAO) with reported interactions are colored. Binding partners that increase PP5 activity are shown in bold and those that initiate degradation of PP5 are underlined; non-proteinaceous partners are italicized. (B) Schematic illustration of TPR-dependent activation of PP5. Under basal conditions, PP5 is autoinhibited. Its active site is covered by the TPR domains (dark gray) that interact with the αJ-helix (purple) and is therefore not accessible for substrates. Binding of activators, such as HSPs, fatty acids, or small molecules, initiates a conformational change of the TPR domains (indicated through color change) and αJ-helix, disrupting their interaction and releasing autoinhibition. This allows access of the substrate to the active site and induced PP5 activity.

Fig. 4.

TPR-domain-dependent activation of PP5. (A) Residues on the surface of PP5 (PDB ID: 1WAO) with reported interactions are colored. Binding partners that increase PP5 activity are shown in bold and those that initiate degradation of PP5 are underlined; non-proteinaceous partners are italicized. (B) Schematic illustration of TPR-dependent activation of PP5. Under basal conditions, PP5 is autoinhibited. Its active site is covered by the TPR domains (dark gray) that interact with the αJ-helix (purple) and is therefore not accessible for substrates. Binding of activators, such as HSPs, fatty acids, or small molecules, initiates a conformational change of the TPR domains (indicated through color change) and αJ-helix, disrupting their interaction and releasing autoinhibition. This allows access of the substrate to the active site and induced PP5 activity.

Therefore, a significant part of research into PP5 biology focuses on its TPR domain-involved activation. PP5 can be released from its autoinhibited state through the interaction of the TPR domain with endogenous molecules, such as heat-shock proteins (HSPs), polyunsaturated fatty acids, or Ca2+/S100 proteins (Fig. 4B) (Yamaguchi et al., 2012; Russell et al., 1999; Haslbeck et al., 2015; Tallima and El Ridi, 2018; Hong et al., 2017). Furthermore, the allosteric PP5 small-molecule activator 2 (P5SA-2) was found to increase PP5 activity, but barely affects substrate binding (D'Arcy et al., 2019; Haslbeck et al., 2015). P5SA-2 binds to the PP5c domain at its interface with the TPR domain, inducing a conformational change of the TPR domains. This leads to an impaired compatibility of the αJ-helix with its binding groove on the TPR domain and finally in the release of autoinhibition and substrate accessibility in PP5 (Hinds and Sánchez, 2008; Haslbeck et al., 2015).

Similar to other PPPs, PP5 activity can further be modulated by PTMs on its catalytic domain. Phosphorylation of PP5 on Thr362 by the kinase CK1δ results in activation of PP5 in the absence of HSP90 in vitro (Dushukyan et al., 2017). PP5 is also subjected to multi-monoubiquitylation on Lys185 and Lys199 by the VHL complex, which leads to its degradation and serves as a control switch to shut off PP5 activity by its degradation (Dushukyan et al., 2017).

PP6 is the third member of the PP2A-like phosphatase subfamily. Nevertheless, PP6 is not dispensable since genetic deletion of the PP6 gene (PPP6C) revealed that PP6 is vital for early embryogenesis (Ye et al., 2015; Ogoh et al., 2016). Analogous to the two other PP2A-like phosphatases, PP6 forms heterotrimeric holoenzymes (Ohama, 2019). The PP6 holoenzyme is composed of one catalytic subunit PP6c, one of three Sit4-associated protein (SAPS1–3) domain-containing scaffold subunits called PPP6R1, PPP6R2 and PPP6R3, and one of the three ankyrin repeat proteins (ANKRD28, ANKRD44 and ANKRD52) that likely serves as its regulatory subunit (Stefansson et al., 2008).

As for PP4, no structure of the PP6 holoenzyme nor any subunit has been resolved yet, and only partial knowledge of the holoenzyme formation is available. It has been shown that the N-terminal region of the scaffolding subunits (SAPS domains) is sufficient for PP6c binding (Twells et al., 2001). Additionally, prior reports from mouse embryonic fibroblasts indicate that ∼75% of PP4c and PP6c are carboxymethylated, which functions as an assembly-directing PTM for PP2A and PP4 (Hwang et al., 2016; Lyons et al., 2021). However, in contrast to PP2A and PP4, loss of methylation does not affect PP6 holoenzyme formation with any of its scaffolding or regulatory subunits (Lyons et al., 2021). Furthermore, PP6 does not stably associate with protein phosphatase methyl esterase 1 (PME-1; also known as PPME1), indicating a carboxymethylation independent regulation of PP6 complex formation (Yabe et al., 2015; Lyons et al., 2021).

In further contrast to PP2Ac, the protein level of PP6c fluctuates greatly, as it is upregulated upon increased cell density in epithelial cell lines (Ohama et al., 2013). This raises the question of how its half-life is regulated. It has been suggested that the interactions of PP6Rs with the catalytic subunit is critical for its protein stability (Wengrod et al., 2015; Watanabe et al., 2018). Furthermore, PP6 has been found to be degraded by p62 (also known as SQSTM1)-dependent autophagy (Fujiwara et al., 2020); this differs from PP2A degradation, which occurs through the ubiquitin-proteasome system (Fujiwara et al., 2020; Ohama et al., 2013; Yabe et al., 2015).

Overall, PP6 remains the least-studied phosphatase of the here discussed PPPs, despite being the most expressed PPP in HeLa cells (Nagaraj et al., 2011). Of note, depending on the substrate, PP6 is even more sensitive to the commonly used PP2A inhibitor okadaic acid (Prickett and Brautigan, 2006), which is often not considered when this inhibitor is used. It is therefore possible that PP6 is in part the cause of the phenotypes induced by okadaic acid that are, however, attributed to other PPPs. In general, when using such natural toxins as PPP inhibitors, one needs to be aware that they are rarely specific (reviewed by Zhang et al., 2021; Fahs et al., 2016; Swingle et al., 2007). However, more insights into the cellular regulatory mechanisms of PP6 are needed to evaluate this potential bias. In summary, although PP6 is frequently observed to play an important role in many pathways (Ohama, 2019), very little is known about its mechanism and regulation. A better understanding of PP6 function might come from insights obtained for PP2A owing to their similarity.

The importance of each PPP for proper cellular function is indisputable. However, many characteristics of PPP function differ from other phosphatases and kinases. Thus, many mechanisms remain so far ambiguous, and specific targeting of individual PPP holoenzyme proteins is still challenging. Nonetheless, considerable improvements in the survey of the human phosphoproteome have revealed the magnitude of sites that are potentially regulated by the PPP family. Furthermore, initial large-scale approaches performed with the major PPPs, PP1 and PP2A, have helped to narrow the gap between site identification and attribution. For instance, it has been demonstrated that PP1c contains intrinsic substrate specificity, which is determined by the B-subunits for PP2A (Hoermann et al., 2020; Kruse et al., 2020). Many recent observations show that SLiM-mediated regulation is common among various aspects of PPP biology. By using SLiMs, PP1c recruits regulatory proteins, which can, but do not have to be, substrates, whereas PP2A holoenzymes (the B-subunits) bind substrate proteins. Interestingly, SLiMs described in this Review (RVxF, LxxIxE and FxxP) change their functional properties drastically upon phosphorylation within their variable positions (x) and at motif-surrounding residues (Nasa et al., 2018; Hertz et al., 2016; Ueki et al., 2019). This indicates an integral layer of regulation, which further fine tunes the SLiM-mediated regulation of PPPs. So far, our understanding of PPP biology is heavily biased towards PP1 and PP2A. Owing to the prevalence of studies addressing them and the variety of their substrates, they are typically assessed more frequently in experiments. However, this bias also has its advantages. Research on PP4 and PP6, for example, benefits greatly from new experimental designs and findings gained from research into PP2A because of their similarity to PP2A. Finally, the understanding of the TPR-driven regulation of the multidomain phosphatase PP5 has a huge potential for its specific targetability. Here, the combination of structural biology, crystallography, phosphoproteomics and chemical biology emerges as a successful approach to better understand PPPs. Thus, we anticipate many meaningful discoveries relating to PPPs in the upcoming years.

We thank the following funding agencies for supporting our work; the German Research Foundation (DFG) within the FOR 2743 (KO 4013/6-1 and HO 1518/13-1) and the CRC 1381 (Project-ID 403222702 – SFB 1381), Germany's Excellence Initiative (DFG, EXC 294 BIOSS), Germany's Excellence Strategy (DFG, CIBSS – EXC-2189 – Project ID 390939984), and the European Research Council with an ERC consolidator grant (#865119) and an ERC starting grant (#336567) to M.K.

Funding

This work was funded by the German Research Foundation (DFG) within the FOR 2743 (KO 4013/6-1) and by the European Research Council with an ERC consolidator grant (#865119).

Afshar
,
K.
,
Werner
,
M. E.
,
Tse
,
Y. C.
,
Glotzer
,
M.
and
Gönczy
,
P.
(
2010
).
Regulation of cortical contractility and spindle positioning by the protein phosphatase 6 PPH-6 in one-cell stage C. elegans embryos
.
Development
137
,
237
-
247
.
Andreeva
,
A. V.
and
Kutuzov
,
M. A.
(
2001
).
PPP family of protein Ser/Thr phosphatases: two distinct branches?
Mol. Biol. Evol.
18
,
448
-
452
.
Ball
,
L. J.
,
Jarchau
,
T.
,
Oschkinat
,
H.
and
Walter
,
U.
(
2002
).
EVH1 domains: structure, function and interactions
.
FEBS Lett.
513
,
45
-
52
.
Baharians
,
Z.
and
Schönthal
,
A. H.
(
1998
).
Autoregulation of protein phosphatase type 2A expression
.
J. Biol. Chem.
273
,
19019
-
19024
.
Blazek
,
M.
,
Santisteban
,
T. S.
,
Zengerle
,
R.
and
Meier
,
M.
(
2015
).
Analysis of fast protein phosphorylation kinetics in single cells on a microfluidic chip
.
Lab. Chip
15
,
726
-
734
.
Bollen
,
M.
,
Peti
,
W.
,
Ragusa
,
M. J.
and
Beullens
,
M.
(
2010
).
The extended PP1 toolkit: designed to create specificity
.
Trends Biochem. Sci.
35
,
450
-
458
.
Brautigan
,
D. L.
(
2013
).
Protein Ser/Thr phosphatases--the ugly ducklings of cell signalling
.
FEBS J.
280
,
324
-
345
.
Brautigan
,
D. L.
and
Shenolikar
,
S.
(
2018
).
Protein serine/threonine phosphatases: keys to unlocking regulators and substrates
.
Annu. Rev. Biochem.
87
,
921
-
964
.
Cao
,
X.
,
Lemaire
,
S.
and
Bollen
,
M.
(
2021
).
Protein phosphatase 1: life-course regulation by SDS22 and Inhibitor-3
.
FEBS J.
289
,
3072
-
3085
.
Ceulemans
,
H.
,
Vulsteke
,
V.
,
de Maeyer
,
M.
,
Tatchell
,
K.
,
Stalmans
,
W.
and
Bollen
,
M.
(
2002
).
Binding of the concave surface of the Sds22 superhelix to the alpha 4/alpha 5/alpha 6-triangle of protein phosphatase-1
.
J. Biol. Chem.
277
,
47331
-
47337
.
Chaklader
,
M.
and
Rothermel
,
B. A.
(
2021
).
Calcineurin in the heart: New horizons for an old friend
.
Cell. Signal.
87
,
110134
.
Chakravarty
,
D.
and
Porter
,
L. L.
(
2022
).
AlphaFold2 fails to predict protein fold switching
.
Protein Sci.
31
,
e4353
.
Chatterjee
,
J.
,
Beullens
,
M.
,
Sukackaite
,
R.
,
Qian
,
J.
,
Lesage
,
B.
,
Hart
,
D. J.
,
Bollen
,
M.
and
Köhn
,
M.
(
2012
).
Development of a peptide that selectively activates protein phosphatase-1 in living cells
.
Angew. Chem. Int. Ed. Engl.
51
,
10054
-
10059
.
Chaudhari
,
M.
,
Thapa
,
N.
,
Ismail
,
H.
,
Chopade
,
S.
,
Caragea
,
D.
,
Köhn
,
M.
,
Newman
,
R. H.
and
Kc
,
D. B.
(
2021
).
DTL-DephosSite: deep transfer learning based approach to predict Dephosphorylation sites
.
Front. Cell Dev. Biol.
9
,
662983
.
Chen
,
R.
,
Rato
,
C.
,
Yan
,
Y.
,
Crespillo-Casado
,
A.
,
Clarke
,
H. J.
,
Harding
,
H. P.
,
Marciniak
,
S. J.
,
Read
,
R. J.
and
Ron
,
D.
(
2015
).
G-actin provides substrate-specificity to eukaryotic initiation factor 2α holophosphatases
.
eLife
4
,
e04871
.
Chen
,
M. J.
,
Dixon
,
J. E.
and
Manning
,
G.
(
2017
).
Genomics and evolution of protein phosphatases
.
Sci. Signal.
10
,
eaag1796
.
Chen
,
P.-H.
,
Hu
,
Z.
,
An
,
E.
,
Okeke
,
I.
,
Zheng
,
S.
,
Luo
,
X.
,
Gong
,
A.
,
Jaime-Figueroa
,
S.
and
Crews
,
C. M.
(
2021
).
Modulation of phosphoprotein activity by phosphorylation targeting chimeras (PhosTACs)
.
ACS Chem. Biol.
16
,
2808
-
2815
.
Cho
,
U. S.
and
Xu
,
W.
(
2007
).
Crystal structure of a protein phosphatase 2A heterotrimeric holoenzyme
.
Nature
445
,
53
-
57
.
Cho
,
M.-H.
,
Wrabl
,
J. O.
,
Taylor
,
J.
and
Hilser
,
V. J.
(
2020
).
Hidden dynamic signatures drive substrate selectivity in the disordered phosphoproteome
.
Proc. Natl. Acad. Sci. U.S.A.
117
,
23606
-
23616
.
Choy
,
M. S.
,
Hieke
,
M.
,
Kumar
,
G. S.
,
Lewis
,
G. R.
,
Gonzalez-DeWhitt
,
K. R.
,
Kessler
,
R. P.
,
Stein
,
B. J.
,
Hessenberger
,
M.
,
Nairn
,
A. C.
,
Peti
,
W.
et al. 
(
2014
).
Understanding the antagonism of retinoblastoma protein dephosphorylation by PNUTS provides insights into the PP1 regulatory code
.
Proc. Natl. Acad. Sci. U.S.A.
111
,
4097
-
4102
.
Choy
,
M. S.
,
Swingle
,
M.
,
D'Arcy
,
B.
,
Abney
,
K.
,
Rusin
,
S. F.
,
Kettenbach
,
A. N.
,
Page
,
R.
,
Honkanen
,
R. E.
and
Peti
,
W.
(
2017
).
PP1:Tautomycetin complex reveals a path toward the development of PP1-specific inhibitors
.
J. Am. Chem. Soc.
139
,
17703
-
17706
.
Choy
,
M. S.
,
Moon
,
T. M.
,
Ravindran
,
R.
,
Bray
,
J. A.
,
Robinson
,
L. C.
,
Archuleta
,
T. L.
,
Shi
,
W.
,
Peti
,
W.
,
Tatchell
,
K.
and
Page
,
R.
(
2019
).
SDS22 selectively recognizes and traps metal-deficient inactive PP1
.
Proc. Natl. Acad. Sci. U.S.A.
116
,
20472
-
20481
.
Cohen
,
P. T. W.
(
2002
).
Protein phosphatase 1--targeted in many directions
.
J. Cell Sci.
115
,
241
-
256
.
Cohen
,
P. T. W.
,
Philp
,
A.
and
Vázquez-Martin
,
C.
(
2005
).
Protein phosphatase 4--from obscurity to vital functions
.
FEBS Lett.
579
,
3278
-
3286
.
Coscia
,
F.
,
Lengyel
,
E.
,
Duraiswamy
,
J.
,
Ashcroft
,
B.
,
Bassani-Sternberg
,
M.
,
Wierer
,
M.
,
Johnson
,
A.
,
Wroblewski
,
K.
,
Montag
,
A.
,
Yamada
,
S. D.
et al. 
(
2018
).
Multi-level proteomics identifies CT45 as a chemosensitivity mediator and immunotherapy target in ovarian cancer
.
Cell
175
,
159
-
170.e16
.
Creamer
,
T. P.
(
2020
).
Calcineurin
.
Cell Commun. Signal.
18
,
137
.
Cundell
,
M. J.
,
Hutter
,
L. H.
,
Nunes Bastos
,
R.
,
Poser
,
E.
,
Holder
,
J.
,
Mohammed
,
S.
,
Novak
,
B.
and
Barr
,
F. A.
(
2016
).
A PP2A-B55 recognition signal controls substrate dephosphorylation kinetics during mitotic exit
.
J. Cell Biol.
214
,
539
-
554
.
Damle
,
N. P.
and
Köhn
,
M.
(
2019
).
The human DEPhOsphorylation database DEPOD: 2019 update
.
Database
2019
,
baz133
.
Danielsson
,
A.
,
Ost
,
A.
,
Nystrom
,
F. H.
and
Strålfors
,
P.
(
2005
).
Attenuation of insulin-stimulated insulin receptor substrate-1 serine 307 phosphorylation in insulin resistance of type 2 diabetes
.
J. Biol. Chem.
280
,
34389
-
34392
.
D'Arcy
,
B. M.
,
Swingle
,
M. R.
,
Papke
,
C. M.
,
Abney
,
K. A.
,
Bouska
,
E. S.
,
Prakash
,
A.
and
Honkanen
,
R. E.
(
2019
).
The antitumor drug LB-100 is a catalytic inhibitor of protein phosphatase 2A (PPP2CA) and 5 (PPP5C) coordinating with the active-site catalytic metals in PPP5C
.
Mol. Cancer Ther.
18
,
556
-
566
.
Dushukyan
,
N.
,
Dunn
,
D. M.
,
Sager
,
R. A.
,
Woodford
,
M. R.
,
Loiselle
,
D. R.
,
Daneshvar
,
M.
,
Baker-Williams
,
A. J.
,
Chisholm
,
J. D.
,
Truman
,
A. W.
,
Vaughan
,
C. K.
et al. 
(
2017
).
Phosphorylation and ubiquitination regulate protein phosphatase 5 activity and its prosurvival role in kidney cancer
.
Cell Rep.
21
,
1883
-
1895
.
Egloff
,
M. P.
,
Johnson
,
D. F.
,
Moorhead
,
G.
,
Cohen
,
P. T.
,
Cohen
,
P.
and
Barford
,
D.
(
1997
).
Structural basis for the recognition of regulatory subunits by the catalytic subunit of protein phosphatase 1
.
EMBO J.
16
,
1876
-
1887
.
Eidenmüller
,
J.
,
Fath
,
T.
,
Maas
,
T.
,
Pool
,
M.
,
Sontag
,
E.
and
Brandt
,
R.
(
2001
).
Phosphorylation-mimicking glutamate clusters in the proline-rich region are sufficient to simulate the functional deficiencies of hyperphosphorylated tau protein
.
Biochem. J.
357
,
759
-
767
.
Fahs
,
S.
,
Lujan
,
P.
and
Köhn
,
M.
(
2016
).
Approaches to study phosphatases
.
ACS Chem. Biol.
11
,
2944
-
2961
.
Fedoryshchak
,
R. O.
,
Přechová
,
M.
,
Butler
,
A. M.
,
Lee
,
R.
,
O'Reilly
,
N.
,
Flynn
,
H. R.
,
Snijders
,
A. P.
,
Eder
,
N.
,
Ultanir
,
S.
,
Mouilleron
,
S.
et al. 
(
2020
).
Molecular basis for substrate specificity of the Phactr1/PP1 phosphatase holoenzyme
.
eLife
9
,
e61509
.
Fischer
,
T. H.
,
Eiringhaus
,
J.
,
Dybkova
,
N.
,
Saadatmand
,
A.
,
Pabel
,
S.
,
Weber
,
S.
,
Wang
,
Y.
,
Köhn
,
M.
,
Tirilomis
,
T.
,
Ljubojevic
,
S.
et al. 
(
2018
).
Activation of protein phosphatase 1 by a selective phosphatase disrupting peptide reduces sarcoplasmic reticulum Ca2+ leak in human heart failure
.
Eur. J. Heart Fail
20
,
1673
-
1685
.
Fowle
,
H.
,
Zhao
,
Z.
and
Graña
,
X.
(
2019
).
PP2A holoenzymes, substrate specificity driving cellular functions and deregulation in cancer
.
Adv. Cancer Res.
144
,
55
-
93
.
Fowle
,
H.
,
Zhao
,
Z.
,
Xu
,
Q.
,
Wasserman
,
J. S.
,
Wang
,
X.
,
Adeyemi
,
M.
,
Feiser
,
F.
,
Kurimchak
,
A. N.
,
Atar
,
D.
,
McEwan
,
B. C.
et al. 
(
2021
).
PP2A/B55α substrate recruitment as defined by the retinoblastoma-related protein p107
.
eLife
10
,
e63181
.
Frohner
,
I. E.
,
Mudrak
,
I.
,
Kronlachner
,
S.
,
Schüchner
,
S.
and
Ogris
,
E.
(
2020
).
Antibodies recognizing the C terminus of PP2A catalytic subunit are unsuitable for evaluating PP2A activity and holoenzyme composition
.
Sci. Signal.
13
,
eaax6490
.
Fujiwara
,
N.
,
Shibutani
,
S.
,
Sakai
,
Y.
,
Watanabe
,
T.
,
Kitabayashi
,
I.
,
Oshima
,
H.
,
Oshima
,
M.
,
Hoshida
,
H.
,
Akada
,
R.
,
Usui
,
T.
et al. 
(
2020
).
Autophagy regulates levels of tumor suppressor enzyme protein phosphatase 6
.
Cancer Sci.
111
,
4371
-
4380
.
Fukuchi
,
S.
,
Hosoda
,
K.
,
Homma
,
K.
,
Gojobori
,
T.
and
Nishikawa
,
K.
(
2011
).
Binary classification of protein molecules into intrinsically disordered and ordered segments
.
BMC Struct. Biol.
11
,
29
.
Gingras
,
A.-C.
,
Caballero
,
M.
,
Zarske
,
M.
,
Sanchez
,
A.
,
Hazbun
,
T. R.
,
Fields
,
S.
,
Sonenberg
,
N.
,
Hafen
,
E.
,
Raught
,
B.
and
Aebersold
,
R.
(
2005
).
A novel, evolutionarily conserved protein phosphatase complex involved in cisplatin sensitivity
.
Mol. Cell. Proteomics
4
,
1725
-
1740
.
Godfrey
,
M.
,
Touati
,
S. A.
,
Kataria
,
M.
,
Jones
,
A.
,
Snijders
,
A. P.
and
Uhlmann
,
F.
(
2017
).
PP2ACdc55 phosphatase imposes ordered cell-cycle phosphorylation by opposing threonine phosphorylation
.
Mol. Cell
65
,
393
-
402.e3
.
Goldberg
,
J.
,
Huang
,
H. B.
,
Kwon
,
Y. G.
,
Greengard
,
P.
,
Nairn
,
A. C.
and
Kuriyan
,
J.
(
1995
).
Three-dimensional structure of the catalytic subunit of protein serine/threonine phosphatase-1
.
Nature
376
,
745
-
753
.
Grundke-Iqbal
,
I.
,
Iqbal
,
K.
,
Tung
,
Y. C.
,
Quinlan
,
M.
,
Wisniewski
,
H. M.
and
Binder
,
L. I.
(
1986
).
Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology
.
Proc. Natl. Acad. Sci. U.S.A.
83
,
4913
-
4917
.
Guccione
,
E.
and
Richard
,
S.
(
2019
).
The regulation, functions and clinical relevance of arginine methylation
.
Nat. Rev. Mol. Cell Biol.
20
,
642
-
657
.
Ham
,
B. M.
,
Jayachandran
,
H.
,
Yang
,
F.
,
Jaitly
,
N.
,
Polpitiya
,
A. D.
,
Monroe
,
M. E.
,
Wang
,
L.
,
Zhao
,
R.
,
Purvine
,
S. O.
,
Livesay
,
E. A.
et al. 
(
2010
).
Novel Ser/Thr protein phosphatase 5 (PP5) regulated targets during DNA damage identified by proteomics analysis
.
J. Proteome Res.
9
,
945
-
953
.
Han
,
X.
,
Gomes
,
J.-E.
,
Birmingham
,
C. L.
,
Pintard
,
L.
,
Sugimoto
,
A.
and
Mains
,
P. E.
(
2009
).
The role of protein phosphatase 4 in regulating microtubule severing in the Caenorhabditis elegans embryo
.
Genetics
181
,
933
-
943
.
Han
,
S.
,
Park
,
J.
and
Lee
,
D.-H.
(
2015
).
Protein DHX38 is a novel inhibitor of protein phosphatase 4
.
Anim. Cells Syst.
19
,
236
-
244
.
Haslbeck
,
V.
,
Drazic
,
A.
,
Eckl
,
J. M.
,
Alte
,
F.
,
Helmuth
,
M.
,
Popowicz
,
G.
,
Schmidt
,
W.
,
Braun
,
F.
,
Weiwad
,
M.
,
Fischer
,
G.
et al. 
(
2015
).
Selective activators of protein phosphatase 5 target the auto-inhibitory mechanism
.
Biosci. Rep.
35
,
e00210
.
Hauseman
,
Z. J.
,
Fodor
,
M.
,
Dhembi
,
A.
,
Viscomi
,
J.
,
Egli
,
D.
,
Bleu
,
M.
,
Katz
,
S.
,
Park
,
E.
,
Jang
,
D. M.
,
Porter
,
K. A.
et al. 
(
2022
).
Structure of the MRAS-SHOC2-PP1C phosphatase complex
.
Nature
609
,
416
-
423
.
Hein
,
J. B.
,
Hertz
,
E. P. T.
,
Garvanska
,
D. H.
,
Kruse
,
T.
and
Nilsson
,
J.
(
2017
).
Distinct kinetics of serine and threonine dephosphorylation are essential for mitosis
.
Nat. Cell Biol.
19
,
1433
-
1440
.
Heroes
,
E.
,
Lesage
,
B.
,
Görnemann
,
J.
,
Beullens
,
M.
,
van Meervelt
,
L.
and
Bollen
,
M.
(
2013
).
The PP1 binding code: a molecular-lego strategy that governs specificity
.
FEBS J.
280
,
584
-
595
.
Heroes
,
E.
,
van der Hoeven
,
G.
,
Choy
,
M. S.
,
Del Garcia
,
J. P.
,
Ferreira
,
M.
,
Nys
,
M.
,
Derua
,
R.
,
Beullens
,
M.
,
Ulens
,
C.
,
Peti
,
W.
et al. 
(
2019
).
Structure-guided exploration of SDS22 interactions with protein phosphatase PP1 and the splicing factor BCLAF1
.
Structure
27
,
507
-
518.e5
.
Hertz
,
E. P. T.
,
Kruse
,
T.
,
Davey
,
N. E.
,
López-Méndez
,
B.
,
Sigurðsson
,
J. O.
,
Montoya
,
G.
,
Olsen
,
J. V.
and
Nilsson
,
J.
(
2016
).
A conserved motif provides binding specificity to the PP2A-B56 Phosphatase
.
Mol. Cell
63
,
686
-
695
.
Hinds
,
T. D.
and
Sánchez
,
E. R.
(
2008
).
Protein phosphatase 5
.
Int. J. Biochem. Cell Biol.
40
,
2358
-
2362
.
Hoermann
,
B.
and
Köhn
,
M.
(
2021
).
Evolutionary crossroads of cell signaling: PP1 and PP2A substrate sites in intrinsically disordered regions
.
Biochem. Soc. Trans.
49
,
1065
-
1074
.
Hoermann
,
B.
,
Kokot
,
T.
,
Helm
,
D.
,
Heinzlmeir
,
S.
,
Chojnacki
,
J. E.
,
Schubert
,
T.
,
Ludwig
,
C.
,
Berteotti
,
A.
,
Kurzawa
,
N.
,
Kuster
,
B.
et al. 
(
2020
).
Dissecting the sequence determinants for dephosphorylation by the catalytic subunits of phosphatases PP1 and PP2A
.
Nat. Commun.
11
,
3583
.
Hong
,
T.-J.
,
Park
,
K.
,
Choi
,
E.-W.
and
Hahn
,
J.-S.
(
2017
).
Ro 90-7501 inhibits PP5 through a novel, TPR-dependent mechanism
.
Biochem. Biophys. Res. Commun.
482
,
215
-
220
.
Huang
,
X.
,
Liu
,
J.
,
Shen
,
T.
,
Meng
,
X.
,
Dou
,
L.
,
Lin
,
Y.
and
Li
,
J.
(
2016
).
Protein phosphatase 4 plays dual roles during cell proliferation
.
Cell Prolif.
49
,
219
-
235
.
Hunter
,
T.
(
1995
).
Protein kinases and phosphatases: The Yin and Yang of protein phosphorylation and signaling
.
Cell
80
,
225
-
236
.
Hwang
,
J.
,
Lee
,
J. A.
and
Pallas
,
D. C.
(
2016
).
Leucine Carboxyl methyltransferase 1 (LCMT-1) methylates protein phosphatase 4 (PP4) and protein phosphatase 6 (PP6) and differentially regulates the stable formation of different PP4 holoenzymes
.
J. Biol. Chem.
291
,
21008
-
21019
.
Isono
,
M.
,
Niimi
,
A.
,
Oike
,
T.
,
Hagiwara
,
Y.
,
Sato
,
H.
,
Sekine
,
R.
,
Yoshida
,
Y.
,
Isobe
,
S.-Y.
,
Obuse
,
C.
,
Nishi
,
R.
et al. 
(
2017
).
BRCA1 Directs the repair pathway to homologous recombination by promoting 53BP1 Dephosphorylation
.
Cell reports
18
,
520
-
532
.
Janssens
,
V.
,
Longin
,
S.
and
Goris
,
J.
(
2008
).
PP2A holoenzyme assembly: in cauda venenum (the sting is in the tail)
.
Trends Biochem. Sci.
33
,
113
-
121
.
Jayadeva
,
G.
,
Kurimchak
,
A.
,
Garriga
,
J.
,
Sotillo
,
E.
,
Davis
,
A. J.
,
Haines
,
D. S.
,
Mumby
,
M.
and
Graña
,
X.
(
2010
).
B55alpha PP2A holoenzymes modulate the phosphorylation status of the retinoblastoma-related protein p107 and its activation
.
J. Biol. Chem.
285
,
29863
-
29873
.
Jeong
,
B.-C.
,
Bae
,
S. J.
,
Ni
,
L.
,
Zhang
,
X.
,
Bai
,
X.-C.
and
Luo
,
X.
(
2021
).
Cryo-EM structure of the Hippo signaling integrator human STRIPAK
.
Nat. Struct. Mol. Biol.
28
,
290
-
299
.
Jong
,
C. J.
,
Merrill
,
R. A.
,
Wilkerson
,
E. M.
,
Herring
,
L. E.
,
Graves
,
L. M.
and
Strack
,
S.
(
2020
).
Reduction of protein phosphatase 2A (PP2A) complexity reveals cellular functions and dephosphorylation motifs of the PP2A/B'δ holoenzyme
.
J. Biol. Chem.
295
,
5654
-
5668
.
Jumper
,
J.
,
Evans
,
R.
,
Pritzel
,
A.
,
Green
,
T.
,
Figurnov
,
M.
,
Ronneberger
,
O.
,
Tunyasuvunakool
,
K.
,
Bates
,
R.
,
Žídek
,
A.
,
Potapenko
,
A.
et al. 
(
2021
).
Highly accurate protein structure prediction with AlphaFold
.
Nature
596
,
583
-
589
.
Junttila
,
M. R.
,
Puustinen
,
P.
,
Niemelä
,
M.
,
Ahola
,
R.
,
Arnold
,
H.
,
Böttzauw
,
T.
,
Ala-aho
,
R.
,
Nielsen
,
C.
,
Ivaska
,
J.
,
Taya
,
Y.
et al. 
(
2007
).
CIP2A inhibits PP2A in human malignancies
.
Cell
130
,
51
-
62
.
Kao
,
G.
,
Tuck
,
S.
,
Baillie
,
D.
and
Sundaram
,
M. V.
(
2004
).
C. elegans SUR-6/PR55 cooperates with LET-92/protein phosphatase 2A and promotes Raf activity independently of inhibitory Akt phosphorylation sites
.
Development
131
,
755
-
765
.
Kauko
,
O.
,
Imanishi
,
S. Y.
,
Kulesskiy
,
E.
,
Yetukuri
,
L.
,
Laajala
,
T. D.
,
Sharma
,
M.
,
Pavic
,
K.
,
Aakula
,
A.
,
Rupp
,
C.
,
Jumppanen
,
M.
et al. 
(
2020
).
Phosphoproteome and drug-response effects mediated by the three protein phosphatase 2A inhibitor proteins CIP2A, SET, and PME-1
.
J. Biol. Chem.
295
,
4194
-
4211
.
Khoury
,
G. A.
,
Baliban
,
R. C.
and
Floudas
,
C. A.
(
2011
).
Proteome-wide post-translational modification statistics: frequency analysis and curation of the swiss-prot database
.
Sci. Rep.
1
,
90
.
Kim
,
J. W.
,
Berrios
,
C.
,
Kim
,
M.
,
Schade
,
A. E.
,
Adelmant
,
G.
,
Yeerna
,
H.
,
Damato
,
E.
,
Iniguez
,
A. B.
,
Florens
,
L.
,
Washburn
,
M. P.
et al. 
(
2020
).
STRIPAK directs PP2A activity toward MAP4K4 to promote oncogenic transformation of human cells
.
eLife
9
,
e53003
.
Kita
,
A.
,
Matsunaga
,
S.
,
Takai
,
A.
,
Kataiwa
,
H.
,
Wakimoto
,
T.
,
Fusetani
,
N.
,
Isobe
,
M.
and
Miki
,
K.
(
2002
).
Crystal structure of the complex between calyculin A and the catalytic subunit of protein phosphatase 1
.
Structure
10
,
715
-
724
.
Kleiman
,
L. B.
,
Maiwald
,
T.
,
Conzelmann
,
H.
,
Lauffenburger
,
D. A.
and
Sorger
,
P. K.
(
2011
).
Rapid phospho-turnover by receptor tyrosine kinases impacts downstream signaling and drug binding
.
Mol. Cell
43
,
723
-
737
.
Köhn
,
M.
(
2020
).
Turn and face the strange: a new view on phosphatases
.
ACS Cent. Sci.
6
,
467
-
477
.
Koike
,
R.
,
Amano
,
M.
,
Kaibuchi
,
K.
and
Ota
,
M.
(
2020
).
Protein kinases phosphorylate long disordered regions in intrinsically disordered proteins
.
Protein Sci.
29
,
564
-
571
.
Kokot
,
T.
,
Hoermann
,
B.
,
Helm
,
D.
,
Chojnacki
,
J. E.
,
Savitski
,
M. M.
and
Köhn
,
M.
(
2022
).
PLDMS: Phosphopeptide Library Dephosphorylation Followed by Mass Spectrometry Analysis to Determine the Specificity of Phosphatases for Dephosphorylation Site Sequences
.
Methods Mol. Biol.
2499
,
43
-
64
.
Krause
,
T.
,
Grote-Wessels
,
S.
,
Balzer
,
F.
,
Boknik
,
P.
,
Gergs
,
U.
,
Kirchhefer
,
U.
,
Buchwalow
,
I. B.
,
Müller
,
F. U.
,
Schmitz
,
W.
and
Neumann
,
J.
(
2018
).
Successful overexpression of wild-type inhibitor-2 of PP1 in cardiovascular cells
.
Naunyn-Schmiedeberg's Arch. Pharmacol.
391
,
859
-
873
.
Kruse
,
T.
,
Gnosa
,
S. P.
,
Nasa
,
I.
,
Garvanska
,
D. H.
,
Hein
,
J. B.
,
Nguyen
,
H.
,
Samsøe–Petersen
,
J.
,
Lopez–Mendez
,
B.
,
Hertz
,
E. P. T.
,
Schwarz
,
J.
et al. 
(
2020
).
Mechanisms of site–specific dephosphorylation and kinase opposition imposed by PP2A regulatory subunits
.
EMBO J.
39
,
e103695
.
Krysiak
,
J.
,
Unger
,
A.
,
Beckendorf
,
L.
,
Hamdani
,
N.
,
von Frieling-Salewsky
,
M.
,
Redfield
,
M. M.
,
Dos Remedios
,
C. G.
,
Sheikh
,
F.
,
Gergs
,
U.
,
Boknik
,
P.
et al. 
(
2018
).
Protein phosphatase 5 regulates titin phosphorylation and function at a sarcomere-associated mechanosensor complex in cardiomyocytes
.
Nat. Commun.
9
,
262
.
Kück
,
U.
,
Radchenko
,
D.
and
Teichert
,
I.
(
2019
).
STRIPAK, a highly conserved signaling complex, controls multiple eukaryotic cellular and developmental processes and is linked with human diseases
.
Biol. Chem.
400
,
1005
-
1022
.
Kurppa
,
K. J.
and
Westermarck
,
J.
(
2020
).
Good guy in bad company: how STRNs convert PP2A into an oncoprotein
.
Cancer Cell
38
,
20
-
22
.
Kwon
,
J. J.
,
Hajian
,
B.
,
Bian
,
Y.
,
Young
,
L. C.
,
Amor
,
A. J.
,
Fuller
,
J. R.
,
Fraley
,
C. V.
,
Sykes
,
A. M.
,
So
,
J.
,
Pan
,
J.
et al. 
(
2022
).
Structure-function analysis of the SHOC2-MRAS-PP1C holophosphatase complex
.
Nature
609
,
408
-
415
.
Laine
,
A.
,
Nagelli
,
S. G.
,
Farrington
,
C.
,
Butt
,
U.
,
Cvrljevic
,
A. N.
,
Vainonen
,
J. P.
,
Feringa
,
F. M.
,
Grönroos
,
T. J.
,
Gautam
,
P.
,
Khan
,
S.
et al. 
(
2021
).
CIP2A Interacts with TopBP1 and Drives Basal-Like Breast Cancer Tumorigenesis
.
Cancer Res.
81
,
4319
-
4331
.
Lee
,
J. A.
and
Pallas
,
D. C.
(
2007
).
Leucine carboxyl methyltransferase-1 is necessary for normal progression through mitosis in mammalian cells
.
J. Biol. Chem.
282
,
30974
-
30984
.
Lee
,
E. Y.
,
Zhang
,
L.
,
Zhao
,
S.
,
Wei
,
Q.
,
Zhang
,
J.
,
Qi
,
Z. Q.
and
Belmonte
,
E. R.
(
1999
).
Phosphorylase phosphatase: new horizons for an old enzyme
.
Front. Biosci.
4
,
D270
-
D285
.
Leonard
,
D.
,
Huang
,
W.
,
Izadmehr
,
S.
,
O'Connor
,
C. M.
,
Wiredja
,
D. D.
,
Wang
,
Z.
,
Zaware
,
N.
,
Chen
,
Y.
,
Schlatzer
,
D. M.
,
Kiselar
,
J.
et al. 
(
2020
).
Selective PP2A enhancement through biased heterotrimer stabilization
.
Cell
181
,
688
-
701.e16
.
Lesage
,
B.
,
Beullens
,
M.
,
Pedelini
,
L.
,
Garcia-Gimeno
,
M. A.
,
Waelkens
,
E.
,
Sanz
,
P.
and
Bollen
,
M.
(
2007
).
A complex of catalytically inactive protein phosphatase-1 sandwiched between Sds22 and inhibitor-3
.
Biochemistry
46
,
8909
-
8919
.
Li
,
X.
,
Wilmanns
,
M.
,
Thornton
,
J.
and
Köhn
,
M.
(
2013
).
Elucidating human phosphatase-substrate networks
.
Sci. Signal.
6
,
rs10
.
Lipinszki
,
Z.
,
Lefevre
,
S.
,
Savoian
,
M. S.
,
Singleton
,
M. R.
,
Glover
,
D. M.
and
Przewloka
,
M. R.
(
2015
).
Centromeric binding and activity of Protein Phosphatase 4
.
Nat. Commun.
6
,
5894
.
Löw
,
C.
,
Quistgaard
,
E. M.
,
Kovermann
,
M.
,
Anandapadamanaban
,
M.
,
Balbach
,
J.
and
Nordlund
,
P.
(
2014
).
Structural basis for PTPA interaction with the invariant C-terminal tail of PP2A
.
Biol. Chem.
395
,
881
-
889
.
Lyons
,
S. P.
,
Jenkins
,
N. P.
,
Nasa
,
I.
,
Choy
,
M. S.
,
Adamo
,
M. E.
,
Page
,
R.
,
Peti
,
W.
,
Moorhead
,
G. B.
and
Kettenbach
,
A. N.
(
2018
).
A quantitative chemical proteomic strategy for profiling phosphoprotein phosphatases from yeast to humans
.
Mol. Cell. Proteomics
17
,
2448
-
2461
.
Lyons
,
S. P.
,
Greiner
,
E. C.
,
Cressey
,
L. E.
,
Adamo
,
M. E.
and
Kettenbach
,
A. N.
(
2021
).
Regulation of PP2A, PP4, and PP6 holoenzyme assembly by carboxyl-terminal methylation
.
Sci. Rep.
11
,
23031
.
Manning
,
G.
,
Whyte
,
D. B.
,
Martinez
,
R.
,
Hunter
,
T.
and
Sudarsanam
,
S.
(
2002
).
The protein kinase complement of the human genome
.
Science
298
,
1912
-
1934
.
Martin-Granados
,
C.
,
Philp
,
A.
,
Oxenham
,
S. K.
,
Prescott
,
A. R.
and
Cohen
,
P. T. W.
(
2008
).
Depletion of protein phosphatase 4 in human cells reveals essential roles in centrosome maturation, cell migration and the regulation of Rho GTPases
.
Int. J. Biochem. Cell Biol.
40
,
2315
-
2332
.
Maynes
,
J. T.
,
Bateman
,
K. S.
,
Cherney
,
M. M.
,
Das
,
A. K.
,
Luu
,
H. A.
,
Holmes
,
C. F.
and
James
,
M. N.
(
2001
).
Crystal structure of the tumor-promoter okadaic acid bound to protein phosphatase-1
.
J. Biol. Chem.
276
,
44078
-
44082
.
Meiselbach
,
H.
,
Sticht
,
H.
and
Enz
,
R.
(
2006
).
Structural analysis of the protein phosphatase 1 docking motif: molecular description of binding specificities identifies interacting proteins
.
Chem. Biol.
13
,
49
-
59
.
Meyerovitch
,
J.
,
Backer
,
J. M.
and
Kahn
,
C. R.
(
1989
).
Hepatic phosphotyrosine phosphatase activity and its alterations in diabetic rats
.
J. Clin. Investig.
84
,
976
-
983
.
Morita
,
K.
,
He
,
S.
,
Nowak
,
R. P.
,
Wang
,
J.
,
Zimmerman
,
M. W.
,
Fu
,
C.
,
Durbin
,
A. D.
,
Martel
,
M. W.
,
Prutsch
,
N.
,
Gray
,
N. S.
et al. 
(
2020
).
Allosteric activators of protein phosphatase 2A display broad antitumor activity mediated by Dephosphorylation of MYBL2
.
Cell
181
,
702
-
715.e20
.
Morita
,
K.
,
He
,
S.
,
Nowak
,
R.P.
,
Wang
,
J.
,
Zimmerman
,
M.W.
,
Fu
,
C.
,
Durbin
,
A.D.
,
Martel
,
M.W.
,
Prutsch
,
N.
,
Gray
,
N.S.
,
Fischer
,
E.S.
and
Look
,
A.T.
(
2022
).
Retraction Notice to: Allosteric activators of protein phosphatase 2A display broad antitumor activity mediated by dephosphorylation of MYBL2
.
Cell
185
,
3058
. .
Nagaraj
,
N.
,
Wisniewski
,
J. R.
,
Geiger
,
T.
,
Cox
,
J.
,
Kircher
,
M.
,
Kelso
,
J.
,
Pääbo
,
S.
and
Mann
,
M.
(
2011
).
Deep proteome and transcriptome mapping of a human cancer cell line
.
Mol. Syst. Biol.
7
,
548
.
Nasa
,
I.
and
Kettenbach
,
A. N.
(
2018
).
Coordination of protein kinase and phosphoprotein phosphatase activities in mitosis
.
Front. Cell Dev. Biol.
6
,
30
.
Nasa
,
I.
,
Rusin
,
S. F.
,
Kettenbach
,
A. N.
and
Moorhead
,
G. B.
(
2018
).
Aurora B opposes PP1 function in mitosis by phosphorylating the conserved PP1-binding RVxF motif in PP1 regulatory proteins
.
Sci. Signal.
11
,
eaai8669
.
Nasa
,
I.
,
Cressey
,
L. E.
,
Kruse
,
T.
,
Hertz
,
E. P. T.
,
Gui
,
J.
,
Graves
,
L. M.
,
Nilsson
,
J.
and
Kettenbach
,
A. N.
(
2020
).
Quantitative kinase and phosphatase profiling reveal that CDK1 phosphorylates PP2Ac to promote mitotic entry
.
Sci. Signal.
13
,
eaba7823
.
Needham
,
E. J.
,
Parker
,
B. L.
,
Burykin
,
T.
,
James
,
D. E.
and
Humphrey
,
S. J.
(
2019
).
Illuminating the dark phosphoproteome
.
Sci. Signal.
12
,
eaau8645
.
Neumann
,
J.
,
Boknik
,
P.
,
Kirchhefer
,
U.
and
Gergs
,
U.
(
2021
).
The role of PP5 and PP2C in cardiac health and disease
.
Cell. Signal.
85
,
110035
.
Nicolaou
,
P.
and
Kranias
,
E. G.
(
2009
).
Role of PP1 in the regulation of Ca cycling in cardiac physiology and pathophysiology
.
Front. Biosci.
14
,
3571
-
3585
.
Nilsson
,
J.
(
2019
).
Protein phosphatases in the regulation of mitosis
.
J. Cell Biol.
218
,
395
-
409
.
Oberoi
,
J.
,
Dunn
,
D. M.
,
Woodford
,
M. R.
,
Mariotti
,
L.
,
Schulman
,
J.
,
Bourboulia
,
D.
,
Mollapour
,
M.
and
Vaughan
,
C. K.
(
2016
).
Structural and functional basis of protein phosphatase 5 substrate specificity
.
Proc. Natl. Acad. Sci. U.S.A.
113
,
9009
-
9014
.
Ochoa
,
D.
,
Jarnuczak
,
A. F.
,
Viéitez
,
C.
,
Gehre
,
M.
,
Soucheray
,
M.
,
Mateus
,
A.
,
Kleefeldt
,
A. A.
,
Hill
,
A.
,
Garcia-Alonso
,
L.
,
Stein
,
F.
et al. 
(
2019
).
The functional landscape of the human phosphoproteome
.
Nat. Biotechnol.
38
,
365
-
373
.
Ogoh
,
H.
,
Tanuma
,
N.
,
Matsui
,
Y.
,
Hayakawa
,
N.
,
Inagaki
,
A.
,
Sumiyoshi
,
M.
,
Momoi
,
Y.
,
Kishimoto
,
A.
,
Suzuki
,
M.
,
Sasaki
,
N.
et al. 
(
2016
).
The protein phosphatase 6 catalytic subunit (Ppp6c) is indispensable for proper post-implantation embryogenesis
.
Mech. Dev.
139
,
1
-
9
.
Ohama
,
T.
(
2019
).
The multiple functions of protein phosphatase 6
.
Biochim. Biophys. Acta Mol. Cell Res.
1866
,
74
-
82
.
Ohama
,
T.
,
Wang
,
L.
,
Griner
,
E. M.
and
Brautigan
,
D. L.
(
2013
).
Protein Ser/Thr phosphatase-6 is required for maintenance of E-cadherin at adherens junctions
.
BMC Cell Biol.
14
,
42
.
Oliveira
,
J.
,
Costa
,
M.
,
de Almeida
,
M. S. C.
,
Da Cruz E Silva
,
O. A. B.
and
Henriques
,
A. G.
(
2017
).
Protein phosphorylation is a key mechanism in Alzheimer's disease
.
J. Alzheimer's Dis.
58
,
953
-
978
.
Olsen
,
J. V.
,
Vermeulen
,
M.
,
Santamaria
,
A.
,
Kumar
,
C.
,
Miller
,
M. L.
,
Jensen
,
L. J.
,
Gnad
,
F.
,
Cox
,
J.
,
Jensen
,
T. S.
,
Nigg
,
E. A.
et al. 
(
2010
).
Quantitative phosphoproteomics reveals widespread full phosphorylation site occupancy during mitosis
.
Sci. Signal.
3
,
ra3
.
Park
,
J.
and
Lee
,
D.-H.
(
2020
).
Functional roles of protein phosphatase 4 in multiple aspects of cellular physiology: a friend and a foe
.
BMB Rep.
53
,
181
-
190
.
Park
,
J.
,
Lee
,
J.
and
Lee
,
D.-H.
(
2019
).
Identification of protein phosphatase 4 inhibitory protein that plays an indispensable role in DNA damage response
.
Mol. Cells
42
,
546
-
556
.
Peti
,
W.
and
Page
,
R.
(
2015
).
Strategies to make protein serine/threonine (PP1, calcineurin) and tyrosine phosphatases (PTP1B) druggable: achieving specificity by targeting substrate and regulatory protein interaction sites
.
Bioorg. Med. Chem.
23
,
2781
-
2785
.
Peti
,
W.
,
Nairn
,
A. C.
and
Page
,
R.
(
2013
).
Structural basis for protein phosphatase 1 regulation and specificity
.
FEBS J.
280
,
596
-
611
.
Prickett
,
T. D.
and
Brautigan
,
D. L.
(
2006
).
The alpha4 regulatory subunit exerts opposing allosteric effects on protein phosphatases PP6 and PP2A
.
J. Biol. Chem.
281
,
30503
-
30511
.
Ragusa
,
M. J.
,
Dancheck
,
B.
,
Critton
,
D. A.
,
Nairn
,
A. C.
,
Page
,
R.
and
Peti
,
W.
(
2010
).
Spinophilin directs protein phosphatase 1 specificity by blocking substrate binding sites
.
Nat. Struct. Mol. Biol.
17
,
459
-
464
.
Ramazi
,
S.
and
Zahiri
,
J.
(
2021
).
Posttranslational modifications in proteins: resources, tools and prediction methods
.
Database
2021
,
baab012
.
Reddy
,
R. J.
,
Gajadhar
,
A. S.
,
Swenson
,
E. J.
,
Rothenberg
,
D. A.
,
Curran
,
T. G.
and
White
,
F. M.
(
2016
).
Early signaling dynamics of the epidermal growth factor receptor
.
Proc. Natl. Acad. Sci. U.S.A.
113
,
3114
-
3119
.
Reither
,
G.
,
Chatterjee
,
J.
,
Beullens
,
M.
,
Bollen
,
M.
,
Schultz
,
C.
and
Köhn
M.
(
2013
).
Chemical activators of protein phosphatase-1 induce calcium release inside intact cells
.
Chem. Biol.
20
,
1179
-
1186
. .
Rogers
,
S.
,
McCloy
,
R.
,
Watkins
,
D. N.
and
Burgess
,
A.
(
2016
).
Mechanisms regulating phosphatase specificity and the removal of individual phosphorylation sites during mitotic exit
.
BioEssays
38
Suppl. 1,
S24
-
S32
.
Rosales
,
K. R.
,
Reid
,
M. A.
,
Yang
,
Y.
,
Tran
,
T. Q.
,
Wang
,
W.-I.
,
Lowman
,
X.
,
Pan
,
M.
and
Kong
,
M.
(
2015
).
TIPRL inhibits protein phosphatase 4 activity and promotes H2AX phosphorylation in the DNA damage response
.
PloS one
10
,
e0145938
.
Ruediger
,
R.
,
van Wart Hood
,
J. E.
,
Mumby
,
M.
and
Walter
,
G.
(
1991
).
Constant expression and activity of protein phosphatase 2A in synchronized cells
.
Mol. Cell. Biol.
11
,
4282
-
4285
.
Rusin
,
S. F.
,
Schlosser
,
K. A.
,
Adamo
,
M. E.
and
Kettenbach
,
A. N.
(
2015
).
Quantitative phosphoproteomics reveals new roles for the protein phosphatase PP6 in mitotic cells
.
Sci. Signal
.
8
,
rs12
.
Russell
,
L. C.
,
Whitt
,
S. R.
,
Chen
,
M. S.
and
Chinkers
,
M.
(
1999
).
Identification of conserved residues required for the binding of a tetratricopeptide repeat domain to heat shock protein 90
.
J. Biol. Chem.
274
,
20060
-
20063
.
Ruvolo
,
P. P.
(
2019
).
Role of protein phosphatases in the cancer microenvironment
.
Biochim. Biophys. Acta Mol. Cell Res.
1866
,
144
-
152
.
Sager
,
R. A.
,
Dushukyan
,
N.
,
Woodford
,
M.
and
Mollapour
,
M.
(
2020
).
Structure and function of the co-chaperone protein phosphatase 5 in cancer
.
Cell Stress Chaperones
25
,
383
-
394
.
Sakamoto
,
K. M.
,
Kim
,
K. B.
,
Kumagai
,
A.
,
Mercurio
,
F.
,
Crews
,
C. M.
and
Deshaies
,
R. J.
(
2001
).
Protacs: chimeric molecules that target proteins to the Skp1-Cullin-F box complex for ubiquitination and degradation
.
Proc. Natl. Acad. Sci. U.S.A.
98
,
8554
-
8559
.
Sandal
,
P.
,
Jong
,
C. J.
,
Merrill
,
R. A.
,
Song
,
J.
and
Strack
,
S.
(
2021
).
Protein phosphatase 2A - structure, function and role in neurodevelopmental disorders
.
J. Cell Sci.
134
,
jcs248187
.
Sangodkar
,
J.
,
Farrington
,
C. C.
,
McClinch
,
K.
,
Galsky
,
M. D.
,
Kastrinsky
,
D. B.
and
Narla
,
G.
(
2016
).
All roads lead to PP2A: exploiting the therapeutic potential of this phosphatase
.
FEBS J.
283
,
1004
-
1024
.
Schüchner
,
S.
,
Behm
,
C.
,
Mudrak
,
I.
and
Ogris
,
E.
(
2020
).
The Myc tag monoclonal antibody 9E10 displays highly variable epitope recognition dependent on neighboring sequence context
.
Sci. Signal.
13
,
eaax9730
.
Seshacharyulu
,
P.
,
Pandey
,
P.
,
Datta
,
K.
and
Batra
,
S. K.
(
2013
).
Phosphatase: PP2A structural importance, regulation and its aberrant expression in cancer
.
Cancer Lett.
335
,
9
-
18
.
Seok
,
S. H.
(
2021
).
Structural insights into protein regulation by phosphorylation and substrate recognition of protein kinases/phosphatases
.
Life
11
,
957
.
Sharma
,
K.
,
D'Souza
,
R. C. J.
,
Tyanova
,
S.
,
Schaab
,
C.
,
Wiśniewski
,
J. R.
,
Cox
,
J.
and
Mann
,
M.
(
2014
).
Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thr-based signaling
.
Cell Rep.
8
,
1583
-
1594
.
Slupe
,
A. M.
,
Merrill
,
R. A.
and
Strack
,
S.
(
2011
).
Determinants for substrate specificity of protein phosphatase 2A
.
Enzyme Res.
2011
,
398751
.
Stefansson
,
B.
,
Ohama
,
T.
,
Daugherty
,
A. E.
and
Brautigan
,
D. L.
(
2008
).
Protein phosphatase 6 regulatory subunits composed of ankyrin repeat domains
.
Biochemistry
47
,
1442
-
1451
.
Su
,
C.
,
Li
,
Z.
,
Cheng
,
J.
,
Li
,
L.
,
Zhong
,
S.
,
Liu
,
L.
,
Zheng
,
Y.
and
Zheng
,
B.
(
2017
).
The protein phosphatase 4 and SMEK1 complex dephosphorylates HYL1 to promote miRNA biogenesis by antagonizing the MAPK cascade in arabidopsis
.
Dev. Cell
41
,
527
-
539.e5
.
Swartz
,
S. Z.
,
Nguyen
,
H. T.
,
McEwan
,
B. C.
,
Adamo
,
M. E.
,
Cheeseman
,
I. M.
and
Kettenbach
,
A. N.
(
2021
).
Selective dephosphorylation by PP2A-B55 directs the meiosis I-meiosis II transition in oocytes
.
eLife
10
,
e70588
.
Swingle
,
M.
,
Ni
,
L.
and
Honkanen
,
R. E.
(
2007
).
Small-molecule inhibitors of ser/thr protein phosphatases: specificity, use and common forms of abuse
.
Method. Mol. Biol.
365
,
23
-
38
.
Tallima
,
H.
and
El Ridi
,
R.
(
2018
).
Arachidonic acid: physiological roles and potential health benefits - a review
.
J. Adv. Res.
11
,
33
-
41
.
Terrak
,
M.
,
Kerff
,
F.
,
Langsetmo
,
K.
,
Tao
,
T.
and
Dominguez
,
R.
(
2004
).
Structural basis of protein phosphatase 1 regulation
.
Nature
429
,
780
-
784
.
Touati
,
S. A.
,
Hofbauer
,
L.
,
Jones
,
A. W.
,
Snijders
,
A. P.
,
Kelly
,
G.
and
Uhlmann
,
F.
(
2019
).
Cdc14 and PP2A phosphatases cooperate to shape phosphoproteome dynamics during mitotic exit
.
Cell Rep.
29
,
2105
-
2119.e4
.
Trebacz
,
M.
,
Wang
,
Y.
,
Makotta
,
L.
,
Henschke
,
L.
and
Köhn
,
M.
(
2020
).
Development of a Photoactivatable Protein Phosphatase-1-Disrupting Peptide
.
J. Org Chem.
85
,
1712
-
1717
.
Twells
,
R. C.
,
Metzker
,
M. L.
,
Brown
,
S. D.
,
Cox
,
R.
,
Garey
,
C.
,
Hammond
,
H.
,
Hey
,
P. J.
,
Levy
,
E.
,
Nakagawa
,
Y.
,
Philips
,
M. S.
et al. 
(
2001
).
The sequence and gene characterization of a 400-kb candidate region for IDDM4 on chromosome 11q13
.
Genomics
72
,
231
-
242
.
Ubersax
,
J. A.
and
Ferrell
,
J. E.
(
2007
).
Mechanisms of specificity in protein phosphorylation
.
Nat. Rev. Mol. Cell Biol.
8
,
530
-
541
.
Ueki
,
Y.
,
Kruse
,
T.
,
Weisser
,
M. B.
,
Sundell
,
G. N.
,
Larsen
,
M. S. Y.
,
Mendez
,
B. L.
,
Jenkins
,
N. P.
,
Garvanska
,
D. H.
,
Cressey
,
L.
,
Zhang
,
G.
et al. 
(
2019
).
A consensus binding motif for the PP4 protein phosphatase
.
Mol. Cell
76
,
953
-
964.e6
.
Ueki
,
Y.
,
Hadders
,
M. A.
,
Weisser
,
M. B.
,
Nasa
,
I.
,
Sotelo–Parrilla
,
P.
,
Cressey
,
L. E.
,
Gupta
,
T.
,
Hertz
,
E. P. T.
,
Kruse
,
T.
,
Montoya
,
G.
et al. 
(
2021
).
A highly conserved pocket on PP2A–B56 is required for hSgo1 binding and cohesion protection during mitosis
.
EMBO Rep.
22
,
e52295
.
Vainonen
,
J. P.
,
Momeny
,
M.
and
Westermarck
,
J.
(
2021
).
Druggable cancer phosphatases
.
Sci. Trans. Med.
13
,
eabe2967
.
Verbinnen
,
I.
,
Ferreira
,
M.
and
Bollen
,
M.
(
2017
).
Biogenesis and activity regulation of protein phosphatase 1
.
Biochem. Soc. Trans.
45
,
89
-
99
.
Virshup
,
D. M.
and
Shenolikar
,
S.
(
2009
).
From promiscuity to precision: protein phosphatases get a makeover
.
Mol. Cell
33
,
537
-
545
.
Vit
,
G.
,
Duro
,
J.
,
Rajendraprasad
,
G.
,
Hertz
,
E. P. T.
,
Holland
,
L. K. K.
,
Weisser
,
M. B.
,
McEwan
,
B. C.
,
Lopez-Mendez
,
B.
,
Sotelo-Parrilla
,
P.
,
Jeyaprakash
,
A. A.
et al. 
(
2022
).
Chemogenetic profiling reveals PP2A-independent cytotoxicity of proposed PP2A activators iHAP1 and DT-061
.
EMBO J.
41
,
e110611
.
Voss
,
M.
,
Campbell
,
K.
,
Saranzewa
,
N.
,
Campbell
,
D. G.
,
Hastie
,
C. J.
,
Peggie
,
M. W.
,
Martin-Granados
,
C.
,
Prescott
,
A. R.
and
Cohen
,
P. T. W.
(
2013
).
Protein phosphatase 4 is phosphorylated and inactivated by Cdk in response to spindle toxins and interacts with γ-tubulin
.
Cell Cycle
12
,
2876
-
2887
.
Wakula
,
P.
,
Beullens
,
M.
,
Ceulemans
,
H.
,
Stalmans
,
W.
and
Bollen
,
M.
(
2003
).
Degeneracy and function of the ubiquitous RVXF motif that mediates binding to protein phosphatase-1
.
J. Biol. Chem.
278
,
18817
-
18823
.
Wang
,
S.
,
Quan
,
L.
,
Li
,
S.
,
You
,
C.
,
Zhang
,
Y.
,
Gao
,
L.
,
Zeng
,
L.
,
Liu
,
L.
,
Qi
,
Y.
,
Mo
,
B.
et al. 
(
2019a
).
The protein phosphatase4 complex promotes transcription and processing of primary microRNAs in arabidopsis
.
Plant Cell
31
,
486
-
501
.
Wang
,
Y.
,
Hoermann
,
B.
,
Pavic
,
K.
,
Trebacz
,
M.
,
Rios
,
P.
and
Köhn
,
M.
(
2019b
).
Interrogating PP1 activity in the mapk pathway with optimized PP1-disrupting peptides
.
Chembiochem
20
,
66
-
71
.
Wang
,
X.
,
Garvanska
,
D. H.
,
Nasa
,
I.
,
Ueki
,
Y.
,
Zhang
,
G.
,
Kettenbach
,
A. N.
,
Peti
,
W.
,
Nilsson
,
J.
and
Page
,
R.
(
2020
).
A dynamic charge-charge interaction modulates PP2A:B56 substrate recruitment
.
eLife
9
,
e55966
.
Watanabe
,
K.
,
Umeda
,
T.
,
Niwa
,
K.
,
Naguro
,
I.
and
Ichijo
,
H.
(
2018
).
A PP6-ASK3 module coordinates the bidirectional cell volume regulation under osmotic stress
.
Cell Rep.
22
,
2809
-
2817
.
Wengrod
,
J.
,
Wang
,
D.
,
Weiss
,
S.
,
Zhong
,
H.
,
Osman
,
I.
and
Gardner
,
L. B.
(
2015
).
Phosphorylation of eIF2α triggered by mTORC1 inhibition and PP6C activation is required for autophagy and is aberrant in PP6C-mutated melanoma
.
Sci. Signal.
8
,
ra27
.
Westermarck
,
J.
and
Neel
,
B. G.
(
2020
).
Piecing together a broken tumor suppressor phosphatase for cancer therapy
.
Cell
181
,
514
-
517
.
Winkler
,
C.
,
de Munter
,
S.
,
van Dessel
,
N.
,
Lesage
,
B.
,
Heroes
,
E.
,
Boens
,
S.
,
Beullens
,
M.
,
van Eynde
,
A.
and
Bollen
,
M.
(
2015
).
The selective inhibition of protein phosphatase-1 results in mitotic catastrophe and impaired tumor growth
.
J. Cell Sci.
128
,
4526
-
4537
.
Wlodarchak
,
N.
and
Xing
,
Y.
(
2016
).
PP2A as a master regulator of the cell cycle
.
Crit. Rev. Biochem. Mol. Biol.
51
,
162
-
184
.
Wright
,
P. E.
and
Dyson
,
H. J.
(
2015
).
Intrinsically disordered proteins in cellular signalling and regulation
.
Nature Reviews. Molecular Cell Biology
16
,
18
-
29
.
Wu
,
C.-G.
,
Chen
,
H.
,
Guo
,
F.
,
Yadav
,
V. K.
,
Mcilwain
,
S. J.
,
Rowse
,
M.
,
Choudhary
,
A.
,
Lin
,
Z.
,
Li
,
Y.
,
Gu
,
T.
et al. 
(
2017
).
PP2A-B’ holoenzyme substrate recognition, regulation and role in cytokinesis
.
Cell Discovery
3
,
17027
.
Wu
,
D.
,
de Wever
,
V.
,
Derua
,
R.
,
Winkler
,
C.
,
Beullens
,
M.
,
van Eynde
,
A.
and
Bollen
,
M.
(
2018
).
A substrate-trapping strategy for protein phosphatase PP1 holoenzymes using hypoactive subunit fusions
.
J. Biol. Chem.
293
,
15152
-
15162
.
Xu
,
Y.
,
Chen
,
Y.
,
Xing
,
Y.
,
Chao
,
Y.
and
Shi
,
Y.
(
2006
).
Structure of the protein phosphatase 2A holoenzyme
.
Cell
127
,
1239
-
1251
.
Xu
,
Y.
,
Chen
,
Y.
,
Zhang
,
P.
,
Jeffrey
,
P. D.
and
Shi
,
Y.
(
2008
).
Structure of a protein phosphatase 2A holoenzyme with B55 subunit
.
Mol. Cell
31
,
873
-
885
.
Yabe
,
R.
,
Miura
,
A.
,
Usui
,
T.
,
Mudrak
,
I.
,
Ogris
,
E.
,
Ohama
,
T.
and
Sato
,
K.
(
2015
).
Protein phosphatase methyl-esterase PME-1 protects protein phosphatase 2A from ubiquitin/proteasome degradation
.
PLoS One
10
,
e0145226
.
Yabe
,
R.
,
Tsuji
,
S.
,
Mochida
,
S.
,
Ikehara
,
T.
,
Usui
,
T.
,
Ohama
,
T.
and
Sato
,
K.
(
2018
).
A stable association with PME-1 may be dispensable for PP2A demethylation - implications for the detection of PP2A methylation and immunoprecipitation
.
FEBS Open Biol.
8
,
1486
-
1496
.
Yamaguchi
,
F.
,
Umeda
,
Y.
,
Shimamoto
,
S.
,
Tsuchiya
,
M.
,
Tokumitsu
,
H.
,
Tokuda
,
M.
and
Kobayashi
,
R.
(
2012
).
S100 proteins modulate protein phosphatase 5 function: a link between CA2+ signal transduction and protein dephosphorylation
.
J. Biol. Chem.
287
,
13787
-
13798
.
Yamazoe
,
S.
,
Tom
,
J.
,
Fu
,
Y.
,
Wu
,
W.
,
Zeng
,
L.
,
Sun
,
C.
,
Liu
,
Q.
,
Lin
,
J.
,
Lin
,
K.
,
Fairbrother
,
W. J.
and
Staben
,
S. T.
(
2020
).
Heterobifunctional molecules induce dephosphorylation of kinases - A proof of concept study
.
J. Med. Chem.
63
,
2807
-
2813
.
Yan
,
Y.
,
Harding
,
H. P.
and
Ron
,
D.
(
2021
).
Higher-order phosphatase-substrate contacts terminate the integrated stress response
.
Nat. Struct. Mol. Biol.
28
,
835
-
846
.
Yang
,
J.
,
Roe
,
S. M.
,
Cliff
,
M. J.
,
Williams
,
M. A.
,
Ladbury
,
J. E.
,
Cohen
,
P. T. W.
and
Barford
,
D.
(
2005
).
Molecular basis for TPR domain-mediated regulation of protein phosphatase 5
.
EMBO J.
24
,
1
-
10
.
Yang
,
X.
,
Yang
,
Y.
,
Sun
,
B.-F.
,
Chen
,
Y.-S.
,
Xu
,
J.-W.
,
Lai
,
W.-Y.
,
Li
,
A.
,
Wang
,
X.
,
Bhattarai
,
D. P.
,
Xiao
,
W.
et al. 
(
2017
).
5-methylcytosine promotes mRNA export - NSUN2 as the methyltransferase and ALYREF as an m5C reader
.
Cell Res.
27
,
606
-
625
.
Ye
,
J.
,
Shi
,
H.
,
Shen
,
Y.
,
Peng
,
C.
,
Liu
,
Y.
,
Li
,
C.
,
Deng
,
K.
,
Geng
,
J.
,
Xu
,
T.
,
Zhuang
,
Y.
et al. 
(
2015
).
PP6 controls T cell development and homeostasis by negatively regulating distal TCR signaling
.
J. Immunol.
194
,
1654
-
1664
.
Youn
,
J.-Y.
,
Dunham
,
W. H.
,
Hong
,
S. J.
,
Knight
,
J. D. R.
,
Bashkurov
,
M.
,
Chen
,
G. I.
,
Bagci
,
H.
,
Rathod
,
B.
,
MacLeod
,
G.
,
Eng
,
S. W. M.
et al. 
(
2018
).
High-density proximity mapping reveals the subcellular organization of mRNA-associated granules and bodies
.
Mol. Cell
69
,
517
-
532.e11
.
Yu
,
X. X.
,
Du
,
X.
,
Moreno
,
C. S.
,
Green
,
R. E.
,
Ogris
,
E.
,
Feng
,
Q.
,
Chou
,
L.
,
McQuoid
,
M. J.
and
Pallas
,
D. C.
(
2001
).
Methylation of the protein phosphatase 2A catalytic subunit is essential for association of Balpha regulatory subunit but not SG2NA, striatin, or polyomavirus middle tumor antigen
.
Mol. Biol. Cell
12
,
185
-
199
.
Zanivan
,
S.
,
Meves
,
A.
,
Behrendt
,
K.
,
Schoof
,
E. M.
,
Neilson
,
L. J.
,
Cox
,
J.
,
Tang
,
H. R.
,
Kalna
,
G.
,
van Ree
,
J. H.
,
van Deursen
,
J. M.
et al. 
(
2013
).
In vivo SILAC-based proteomics reveals phosphoproteome changes during mouse skin carcinogenesis
.
Cell reports
3
,
552
-
566
.
Zarin
,
T.
,
Strome
,
B.
,
Peng
,
G.
,
Pritišanac
,
I.
,
Forman-Kay
,
J. D.
and
Moses
,
A. M.
(
2021
).
Identifying molecular features that are associated with biological function of intrinsically disordered protein regions
.
eLife
10
,
e60220
.
Zhang
,
M.
,
Yogesha
,
S. D.
,
Mayfield
,
J. E.
,
Gill
,
G. N.
and
Zhang
,
Y.
(
2013
).
Viewing serine/threonine protein phosphatases through the eyes of drug designers
.
FEBS J.
280
,
4739
-
4760
.
Zhang
,
Q.
,
Fan
,
Z.
,
Zhang
,
L.
,
You
,
Q.
and
Wang
,
L.
(
2021
).
Strategies for targeting serine/threonine protein phosphatases with small molecules in cancer
.
J. Med. Chem.
64
,
8916
-
8938
.
Zhao
,
Z.
,
Kurimchak
,
A.
,
Nikonova
,
A. S.
,
Feiser
,
F.
,
Wasserman
,
J. S.
,
Fowle
,
H.
,
Varughese
,
T.
,
Connors
,
M.
,
Johnson
,
K.
,
Makhov
,
P.
et al. 
(
2019
).
PPP2R2A prostate cancer haploinsufficiency is associated with worse prognosis and a high vulnerability to B55α/PP2A reconstitution that triggers centrosome destabilization
.
Oncogenesis
8
,
72
.
Zheng
,
H.
,
Qi
,
Y.
,
Hu
,
S.
,
Cao
,
X.
,
Xu
,
C.
,
Yin
,
Z.
,
Chen
,
X.
,
Li
,
Y.
,
Liu
,
W.
,
Li
,
J.
et al. 
(
2020
).
Identification of Integrator-PP2A complex (INTAC), an RNA polymerase II phosphatase
.
Science)
370
,
eabb5872
.

Competing interests

The authors declare no competing or financial interests.