Skip to main content

REVIEW article

Front. Cell Dev. Biol., 01 December 2022
Sec. Morphogenesis and Patterning
Volume 10 - 2022 | https://doi.org/10.3389/fcell.2022.963608

The unexpected versatility of ALP/Enigma family proteins

www.frontiersin.orgLucas A. B. Fisher www.frontiersin.orgFrieder Schöck*
  • Department of Biology, McGill University, Montreal, QC, Canada

One of the most intriguing features of multicellular animals is their ability to move. On a cellular level, this is accomplished by the rearrangement and reorganization of the cytoskeleton, a dynamic network of filamentous proteins which provides stability and structure in a stationary context, but also facilitates directed movement by contracting. The ALP/Enigma family proteins are a diverse group of docking proteins found in numerous cellular milieus and facilitate these processes among others. In vertebrates, they are characterized by having a PDZ domain in combination with one or three LIM domains. The family is comprised of CLP-36 (PDLIM1), Mystique (PDLIM2), ALP (PDLIM3), RIL (PDLIM4), ENH (PDLIM5), ZASP (PDLIM6), and Enigma (PDLIM7). In this review, we will outline the evolution and function of their protein domains which confers their versatility. Additionally, we highlight their role in different cellular environments, focusing specifically on recent advances in muscle research using Drosophila as a model organism. Finally, we show the relevance of this protein family to human myopathies and the development of muscle-related diseases.

Introduction

Animal locomotion is one of the most fascinating mechanisms in nature. Evading an enemy, foraging for food, finding a mate; these fundamental processes of life all require locomotory abilities. On a cellular level, this is accomplished by the rearrangement and reorganization of the cytoskeleton, a dynamic network of filamentous proteins which provides stability and structure in a stationary context, but also facilitates directed movement by contracting. All animal cells require regulation of the cytoskeleton and their dynamics. This regulation is facilitated by numerous different classes of proteins, such as crosslinkers, kinases, docking proteins and more.

The ALP/Enigma family proteins are able to bind to both cytoskeletal and nuclear proteins and interact with a number of regulatory proteins (reviewed in Krcmery et al., 2010). The family is characterized by having a PDZ (post synaptic density protein, Drosophila discs large, and zonula occludens-1 protein) domain, LIM (Lin11, Isl-1, Mec-3) domains, a ZM (Zasp motif) domain, and an AM (ALP motif) domain (Koch et al., 2012). While the evolution of PDZ and LIM domains predates metazoans, Enigma proteins with a combination of one N-terminal PDZ domain, an AM domain, and three C-terminal LIM domains originated in the stem lineage of Metazoa, and later gave rise through duplication and recombination to both the ALP and combined ALP/Enigma subclasses in the stem of Bilateria (Figure 1). ALP proteins have a PDZ domain followed by a single, but different LIM domain. Combined ALP/Enigma proteins evolved in certain groups of Bilateria like Ecdysozoa and consist of one PDZ domain followed by one ALP-like LIM domain and three C-terminal Enigma-like LIM domains (te Velthuis et al., 2007; Koch et al., 2012) (Figure 1). Drosophila also has family members that have lost LIM domains entirely (González-Morales et al., 2019a) (Figure 1). Finally, the ZM domain is only found in Bilateria (González-Morales et al., 2019b). Interestingly, all members of the family share a portion of the AM domain, first described in te Velthuis et al., 2007. It was previously believed that this motif only belonged to the ALP protein subfamily, but it has now been discovered to be present in all members of the ALP/Enigma family (Koch et al., 2012). This, together with the order of domains, reinforces their familial evolutionary relationship and indicates that ALP/Enigma members shared a metazoan common ancestor.

FIGURE 1
www.frontiersin.org

FIGURE 1. Origin and distribution of ALP/Enigma proteins in the metazoan tree. ALP/Enigma proteins first evolved in metazoans as evidenced by single Enigma-like proteins found for example in the ancient phyla of Porifera (the sponge Amphimedon) and Cnidaria (the sea anemone Nematostella). In Bilateria, duplication and recombination gave rise to ALP-like proteins with a single, but different LIM domain, but also to combined ALP/Enigma proteins with four LIM domains (e.g. in Drosophila and C. elegans), as well as proteins without LIM domains (Zasp66 and Zasp67 in Drosophila). The ZM domain first evolved in the ancestor of Bilateria. Evolutionary tree with representative species and the ALP/Enigma proteins with their respective proteins domains (PDZ, ZM, LIM) are shown. Arrows indicate the stem lineage where a particular group of ALP/Enigma proteins originated.

In humans, the ALP/Enigma family is comprised of CLP-36 (PDLIM1), Mystique (PDLIM2), ALP (PDLIM3), RIL (PDLIM4), ENH (PDLIM5), ZASP (PDLIM6), and Enigma (PDLIM7) (Figure 1). The first four members belong to the ALP subfamily, while the last three members belong to the Enigma subfamily.

Here we will provide an update of the current research on proteins of the ALP/Enigma family, focusing on their cytoskeletal functions in Drosophila and vertebrates.

The structure and function ofALP/Enigma family protein domains

ALP/Enigma proteins have developed a wide range of functions in the cell, and are able to bind a myriad of different proteins. This is accomplished by the combination of PDZ and LIM protein domains which on one hand can facilitate interaction with the cytoskeleton either directly or indirectly through the help of other proteins such as a-actinin, while also being able to interact with different cytoskeletal regulatory proteins (Table 1). Some members of this family also contain a muscle-specific ZM domain. Although not much is known of this domain, recent research has uncovered its importance in muscle development and structure, which will be a focus of discussion later in this review. The AM domain is not further discussed here because too little is known about it at the present time (Koch et al., 2012). Examining the function of these domains gives insight into how ALP/Enigma proteins developed their versatility with regards to cytoskeletal dynamics.

TABLE 1
www.frontiersin.org

TABLE 1. Interaction partners of the ALP/Enigma family proteins.

The PDZ domain and the key to versatility

The PDZ domain is a common structural domain found in most organisms, and one of the most prevalent interaction modules found in humans (reviewed in Harris and Lim 2001 and Luck et al., 2012). It is composed of six β-strands, capped by one long and one short α-helix (reviewed in Lee and Zheng 2010). Classically, PDZ domains have been organized into three unique classes based on their C-terminal binding motif; class I, which bind a S/T-X-Φ motif (where Φ is a hydrophobic amino acid and X is any amino acid), class II which bind a Φ-X-Φ motif, and class III which recognize D/E-X-Φ. There are also non-canonical PDZ motifs that can recognize internal sequences of target proteins, as well as phospholipids (Lenfant et al., 2010; reviewed in Gallardo et al., 2010). However, this three-pronged, narrow classification has been shown to be simplistic, and in reality, the PDZ binding specificity is often mediated by more than just the terminal 3-4 residues. Recent studies have shown that most PDZ domain binding sites are achieved through interactions with residues comprising the last seven amino acids, and that the PDZ domains can actually be grouped into 16 distinct specificity classes (Tonikian et al., 2008).

To further complicate things, there are other factors that determine PDZ binding cleft specificity. Indirect residue interactions (Ernst et al., 2014) and sequence context (defined as extensions directly upstream or downstream of the PDZ domain) (reviewed in Wang et al., 2010 and Luck et al., 2012) can be important for interaction with ligands. This is evident in members of the ALP/Enigma family. Zasp52, the Drosophila homolog of ZASP, has been shown to bind α-actinin, and thus be a core component of the Z-disc. It was previously believed that the interaction was mediated by the PDZ domain alone (Katzemich et al., 2013). However, recent in vitro biochemical data has revealed that it is not only the PDZ domain, but also a 20–50 amino acid C-terminal extension that mediates this interaction (Liao et al., 2016).

PDZ domains have also been shown to be robust to mutational load during evolution. Studying PDZ domain evolution in vitro using peptide phage display libraries has demonstrated that model PDZ domains are often still able to bind C-terminal peptides after undergoing single point mutations (Tonikian et al., 2008). They are “hardwired” for ligand binding, and their functional properties could permit the rapid evolution of a protein interaction network (Ernst et al., 2009). Additionally, evolved synthetic domains bind their corresponding ligands with higher affinity than reference domains that were not evolved to do so, but interestingly, in comparison to unevolved synthetic domains, they do so with lower specificity (Ernst et al., 2010). This further validates the inherent ability for the PDZ domain to bind cognate partners and its robustness to mutation during evolution.

Recently, insights into the evolutionary paths of PDZ domains have been elucidated which has direct relevance for the ALP/Enigma family. Teyra et al. (2019), using peptide phage display, mapped all possible mutational transitions between the Erbin PDZ domain, a canonical type I domain, and a synthetic Erbin-PDZ variant E-14, which exhibits atypical specificity and shows strong resemblance to the PDZ domain of PDLIM4, a member of the ALP/Enigma family. It was discovered that three substitutions alone conferred two distinct binding specificities, one similar to Erbin-PDZ and the other to that of E-14/PDLIM4, and that four or more substitutions was able to completely convert the binding profile of the variant to that of E-14/PDLIM4. All other members of the ALP/Enigma family demonstrate class I canonical binding (Au et al., 2004; Kalyoncu et al., 2010) including PDLIM3, the closest evolutionary relative to PDLIM4 in regard to the PDZ domain (te Velthuis et al., 2007). Their research showcases that minimal molecular changes in a binding site can drastically modify binding specificity, and provides an understanding of how the PDZ domain of the ALP/Enigma common ancestor could have developed a non-canonical binding specificity during evolution.

This demonstrates the versatility that can be conferred by the inclusion of the PDZ domain in protein structures. The PDZ domain can not only evolve vastly different binding specificities during evolution because of its inherent functional plasticity, but even structurally similar PDZ domain sequences can bind vastly different ligands (Ernst et al., 2009) (Table 1). Moreover, sequence context can often dictate new specificities, which continues to increase the number of protein-protein interactions possible. This explains how the ALP/Enigma family of proteins are able to interact with numerous different regulatory proteins, and how they have become crucial to vastly different processes in the cell.

The LIM domain and mechanosensitive localization to the cytoskeleton

The LIM domain is less well understood than the PDZ domain, but it also boasts an amazing diversity in binding specificities. LIM domains are cysteine and histidine-rich zinc finger protein binding interfaces with the classical consensus sequence of CX2CX16-23HX2CX2CX2CX16-21CX2(C/H/D) (where X denotes any amino acid) and are roughly 50–65 amino acids in length (reviewed in Kadrmas and Beckerle 2004). LIM domains are especially difficult to classify, because the invariant cysteines required to form the zinc finger motif give the false impression of high evolutionary conservation. In reality, LIM domain sequences show low sequence conservation, and are promiscuous in their binding nature (Koch et al., 2012). During their evolution, LIM domains underwent a rapid expansion and burst of promiscuity in the stem lineage of Metazoa, likely having an important contribution in the development of animal multicellularity (Koch et al., 2012).

As for the function of LIM domains, they have been found to bind cytoskeletal proteins and are prominent members of integrin adhesion sites. Integrins transduce mechanical forces from the cellular matrix into biochemical signals in cells. This is achieved via the adhesome, a multiprotein complex composed of cytoskeletal proteins, adaptor proteins and numerous enzymes, which are recruited to integrin adhesion sites. Integrins have no catalytic activity themselves and are incapable of binding F-actin, thus it is necessary to recruit other proteins to the adhesome in order to translate the external forces recognized by the integrin receptors. Each member of the ALP/Enigma family have been shown to be implicated in integrin adhesion sites (Schiller et al., 2011; Bouaouina et al., 2012). Interestingly, they are part of a group of proteins that appear to be recruited to these sites in a myosin-II-dependent manner, suggesting a possible role of LIM domain proteins as tension sensors (Schiller et al., 2011).

Recently, large strides have been made to further understand this recruitment process. Two studies (Sun et al., 2020; Winkelman et al., 2020) uncovered a mechanism by which LIM domains recognize and adhere to the cytoskeleton. LIM domain proteins were found to recognize stress fiber strain sites, areas of stress fibers that are undergoing mechanical stress. This mechanism is accomplished by the LIM domains themselves, and it is facilitated via multiple domains working in tandem separated by pre-set linker sequences of precise length acting as a “ruler.” When the linker sequences are increased, stress fiber strain site binding is abrogated (Winkelman et al., 2020). Additionally, there is an increase in affinity to the stress fiber strain sites with an increase in the number of LIM domains (Winkelman et al., 2020). Interestingly, chimera experiments demonstrated that the LIM domains are functionally swappable (Sun et al., 2020). All of this has relevance for the Enigma subfamily of proteins, which have 3 LIM domains in tandem. This mechanism is conserved in these proteins and contributes to their localization to integrin adhesion sites (Winkelman et al., 2020). However, it is important to note that there are likely other modes by which LIM domains can engage F-actin (Sun et al., 2020), which does not preclude ALP subfamily proteins from interacting with stress fibers. Sun et al. (2020) found that ALP family members (PDLIM1 and PDLIM2) were still recruited to strain sites, but were recruited at a substantially later time point, and their recruitment was associated with an accumulation of α-actinin. This demonstrates that although ALP subfamily members only have one LIM domain, they are still able to reach the stress fiber strain sites in another manner, perhaps mediated by their interaction with α-actinin.

The stress fiber strain site recognition mechanism is highly conserved, as demonstrated by its presence in the fission yeast protein paxillin-like 1 (Pxl1). Winkelman et al. (2020) hypothesize that this mechanosensing mechanism emerged via a duplication and divergence event of an ancestral CRP-like LIM domain. CRP-like proteins are found in plants and are able to bind unstrained actin filaments via tandem LIM domains (Thomas et al., 2007). The divergence of the CRP-like ancestral molecule conferred new specificity for strained actin filaments and later underwent an expansion in the metazoan stem lineage (Winkelman et al., 2020).

LIM domain proteins are critical to the ALP/Enigma proteins. They enable the proteins of the family to recognize stressed cytoskeletal areas, and interact with a slew of different proteins (Table 1). In the context of Cypher/Zasp, LIM domains bind protein kinases (PKC), kinase anchoring proteins (AKAP) and contribute to self-interaction, which will be elaborated upon later (González-Morales et al., 2019b). For members of the Enigma subfamily, having multiple LIM domains in tandem not only increases binding to the actin stress fibres, but can also be used to bind several different proteins at once, highlighting the versatility the proteins can have in regards to protein-protein interactions. In the next section we will finish up the important domains of the ALP/Enigma family with the ZM domain, a domain less well studied than the other two, but with great leaps recently made in uncovering its function.

The muscle-specific ZM domain: The dark horse

Almost nothing is known about the ZM domain, despite many disease-causing mutations being found within this short motif. The ZM motif is found in Drosophila Zasp52, human ZASP, as well as the human ALP proteins PDLIM1 and PDLIM3, which indicates that this domain was likely present in the ancestor before the splitting of subfamilies (Koch et al., 2012). Whereas PDZ domains occur in all branches of life, and LIM domains occur in all eukaryotes, the more recently evolved ZM domain is restricted to Bilateria, higher eukaryotes with canonical sarcomere structure (González-Morales et al., 2019b). This suggests muscle-specific functions, and the first well-documented function of the ZM domain is sarcomere width control, which will be described in a later section.

Limited structural studies like NMR have so far not detected a structure for this 26 amino acid motif, indicating that it could be part of a low-complexity/disordered region (Klaavuniemi et al., 2009; Watts et al., 2017). It has been proposed that the ZM domain is needed for protein-protein interactions with α-actinin, although this interaction is ambiguous, with some studies reporting binding and others not. For example, for both ALP and ZASP there is evidence demonstrating that the ZM domain is necessary for optimal interaction with α-actinin (Klaavuniemi et al., 2004; Martinelli et al., 2014). This could also be the case for CLP36, which contains similar interaction sites for α-actinin (Klaavuniemi et al., 2004). However, other studies, such as Liao et al. (2016) and Lin et al. (2014) found that the ZM domain was not necessary for this interaction, attributing the α-actinin binding to the PDZ or extended PDZ domain region.

In the case of ALP, the ZM domain is necessary for proper recruitment to the Z-disc (Klaavuniemi et al., 2004), and has been proposed to cause the symptoms of myotonic dystrophy type 1 because of faulty alternative splicing events that result in the loss of this domain (Ohsawa et al., 2011). For ZASP, the ZM domain is also necessary for proper recruitment to the Z-disc. Point mutations in this region do not seem to have effects on this phenomenon. The ZM domain mutations A165V and A171T, which are involved in myopathies, have been shown to have no effect on actin binding (Watts et al., 2017), nor on proper recruitment to the Z-disc (Klaavuniemi and Ylänne 2006). However, these mutations do impair binding to Ankrd2, a mechanosensing protein involved in regulation of gene expression and muscle differentiation, as well as α-actinin, highlighting possible reasons for development of muscle related diseases (Martinelli et al., 2014). Additionally, the importance of the ZM domain in determining sarcomere size discussed later suggests that these mutations could be implicated in sarcomere size control and development. Further research needs to be carried out to clarify the role of ZM mutations in sarcomere structure and regulation.

We will now briefly touch on stretches of amino acids found in the ALP/Enigma family that have no known purpose at the moment, but are present in larger isoforms in the intervening sequences between conserved domains. Using ZASP as an example, we will examine what these regions mean for the proteins of this family, and briefly discuss the burgeoning research into intrinsically disordered proteins.

ZASP and the disordered regions of the ALP/Enigma subfamily

Intrinsically disordered protein regions are found between foldable domains and do not form a stable structure, yet are still able to carry out biological functions in a cellular environment (reviewed in Oldfield and Dunker 2014). Many of the ALP/Enigma proteins harbor interactors through binding of the interdomain area, highlighting the importance of these disordered regions (Table 1). In the case of ZASP, there are numerous isoforms that contain disordered regions.

In Drosophila, there are 22 isoforms of Zasp52. Of the known isoforms, the largest is roughly 240 kDa in size and is found exclusively in the indirect flight muscles (IFM) of adults (Chechenova et al., 2013). It contains an enormous stretch of roughly 1700 amino acids in between the first and second LIM domain that corresponds to an intrinsically disordered region. RNAi knockdown of these long Zasp52 isoform cannot be rescued by a transgene containing only PDZ, ZM and LIM domains (Liao et al., 2016). This data suggests that the disordered region contributes to proper structure and function of the IFMs.

In mice, some ZASP isoforms also have different regions and lengths depending on their localization. For example, some isoforms of ZASP contain tissue-specific disordered regions based on their localization pattern in cardiac or skeletal muscle (Huang et al., 2003). This indicates tissue-specific importance of these disordered regions. Additionally, some of the larger isoforms contain a homologous disordered region found before the 3 LIM domains that is necessary for proper structure and function, leading to myopathic phenotypes when lost (Huang et al., 2003). Furthermore, we already know these regions to be important for human disease. There are multiple mutations found within the linker sequences between the PDZ and LIM domains causing dilated cardiomyopathy and left ventricular non-compaction (Vatta et al., 2003). All of these studies demonstrate the importance of the ZASP intrinsically disordered regions in aberrant muscle phenotypes. Further research will need to be carried out in order to elucidate the role of these non-conserved regions in muscular structure and development, and the mechanism by which they complement the PDZ, LIM and ZM domains of the ALP/Enigma proteins to carry out cellular functions.

The many functions of the ALP/Enigma proteins

The ALP/Enigma proteins participate in a wide array of cellular processes. For one, the ALP/Enigma proteins have been shown to be implicated in many different molecular mechanisms in the nervous system including neuritogenesis (Ohno et al., 2009; Hasegawa et al., 2010) and dendrite formation (Iida et al., 2009; Herrick et al., 2010). Additionally, and perhaps in consequence of this, there have been links of multiple members of the family with neurological diseases such as bipolar disorder, schizophrenia, attention deficit/hyperactivity disorder and Alzheimer’s disease (Kato et al., 2005; Li et al., 2008; Wang et al., 2012; Lee et al., 2015; Moselhy et al., 2015) and even some experimental evidence of ENH expression being related to mood disorders in mice (Horiuchi et al., 2013; Lu et al., 2020). ALP/Enigma proteins are also associated with tumor invasiveness. Almost all of the members of the family have been shown to be involved in cancer-related regulatory dynamics (PDLIM1: Liu et al., 2014; Chen et al., 2015; PDLIM2: Qu et al., 2010; Bowe et al., 2014; PDLIM3: Stein et al., 2010; Katkoori et al., 2012; PDLIM4: Vanaja et al., 2009; Jia et al., 2019; PDLIM5: Yan et al., 2015; Liu et al., 2017; PDLIM7: Kales et al., 2014; Klein et al., 2018). This is particularly the case for PDLIM1 and PDLIM2, with their involvement in cancer being reviewed recently in Zhou et al. (2021) and Guo and Qu (2021). ALP/Enigma family members are also involved in hemostasis (Bozulic et al., 2007; Gupta et al., 2012; Krcmery et al., 2013; Urban et al., 2016), immune and inflammatory responses (Qu et al., 2012; Ono et al., 2015; Fu et al., 2019; Joyce et al., 2019; Yoo et al., 2019) and bone morphogenesis (Strohbach et al., 2008; Lin et al., 2010; Liu H et al., 2014), demonstrating the versatility of this protein family. The functional diversity of the ALP/Enigma proteins cannot be overstated, however, here we focus on their roles in muscle structure and development.

ALP/Enigma family proteins in a muscle context

Muscles are made up of myofibrils, which are in turn made up of sarcomeres, the smallest contractile unit of the muscle cell (Figure 2A). The sarcomere contains a stereotypical structure; each unit is delineated by the Z-disc, a multiprotein structure which anchors the actin thin filaments. These are antiparallel to the myosin thick filaments, which are anchored at the M-line, and it is the sliding motion of the myosin thick filaments over the thin filaments that causes contraction. This contraction is heavily dependent on cytoskeletal dynamics, and therefore ALP/Enigma proteins are naturally implicated in this process.

FIGURE 2
www.frontiersin.org

FIGURE 2. The sarcomere and Zasp52 during Z-disc growth (A) Structure of the sarcomere. The myosin thick filaments are linked at the M-line, and the actin thin filaments are anchored at the Z-disc. (B) Process uncovered by González-Morales et al. (2019b). During early Z-disc growth, the “growing” isoform (Zasp52) predominates and allows interaction with other Zasp proteins through the ZM/LIM interaction. During late Z-disc growth, the “blocking” isoforms with no LIM domains predominate (Zasp66 and Zasp67), which inhibits further growth of the Z-disc.

Many of the ALP/Enigma proteins have critical roles in a muscle context. In a landmark study, Pashmforoush et al. demonstrated the role of ALP in the development of dilated cardiomyopathy through its interaction with α-actinin and stabilization of actin filament structure (Pashmforoush et al., 2001). Since then, many more studies have elucidated the role of ALP/Enigma members in muscle structure stability. In C. elegans, the sole ALP/Enigma protein ALP-1 stabilizes the actin filament structure (Han and Beckerle 2009). In regards to cardiac structural integrity, Enigma has been shown to cause valve dysfunction in both zebrafish and mouse heart models (Camarata et al., 2010; Krcmery et al., 2013).

ALP/Enigma proteins also promote differentiation of muscle cells, as well as cardiac remodelling. Recently for example, ALP and ENH have been shown to promote differentiation and proliferation of satellite cells in chicken skeletal muscle, with ALP being regulated by miRNAs (Yin et al., 2020; He et al., 2021). ENH is also involved in the regulation and remodelling of rodent hearts, with different splice variants leading to reduced or enhanced ventricular cardiomyocyte hypertrophy, in addition to being the target of miRNA silencing which promotes cardiomyocyte hypertrophy (Yamazaki et al., 2010; Bang et al., 2014). Moreover, Enigma has been found to be a target of the E3 ubiquitin ligase Nedd4-1 causing the development of muscle atrophy when degraded (D’Cruz et al., 2016). Members of the ALP/Enigma family are therefore involved in proliferation, differentiation, and maintenance of proper structure of muscle cells.

Perhaps the most prevalent and well researched ALP/Enigma protein in muscles is ZASP/Cypher. ZASP (Z-disc associated alternatively spliced protein) is a core component of the Z-disc. Ablation of ZASP (Cypher) in mouse heart muscles causes development of dilated cardiomyopathy with premature death (Zheng et al., 2008). ZASP has also been shown to be involved in myofibrillar myopathies and cardiomyopathies in humans (Arimura et al., 2009; von Nadelstadh et al., 2009; Lin et al., 2014). Therefore it is of great importance to understand binding partners, dynamics and the roles of ZASP in development. Recently, significant insights have been gained using Drosophila melanogaster as a model organism. The indirect flight muscles (IFM) of Drosophila have strong homology to human sarcomeres (reviewed in Lemke and Schnorrer 2017), thus making them an ideal model to study sarcomeric proteins. The next section will focus on recent advances undertaken in the Drosophila model system, and the implication this has for human muscle-related diseases.

From insects to humans: Zasp52 and myofibril development

Zasp52, the Drosophila ZASP ortholog, is necessary for a myriad of processes in muscle cells. For starters, Zasp52 is essential for the maintenance and development of functional integrin adhesion sites, which is integral to cell spreading and the development of myotendinous junctions (Jani and Schöck, 2007). Moreover, Zasp52 has been shown to be required for integrin activation as well. Zasp52 activates integrins in dual color flow cytometric assays, and Zasp52 mutants can be rescued by talin overexpression, the main activator of integrins (Bouaouina et al., 2012). More importantly, however, in vivo fluorescent recovery after photobleaching (FRAP) experiments have demonstrated that integrins are more mobile in Zasp52 mutant embryos and talin mutants during later stages of Drosophila embryo development compared to their wild-type counterparts (Bouaouina et al., 2012), which is indicative of muscle detachment. Mature myofibrils are more static in their integrin adhesion sites in order to resist contractile forces (Yuan et al., 2010), and therefore this highlights the importance of Zasp52 as a regulator of cell signalling during contraction. Without functional integrin adhesion sites, muscle cells are not able to properly bind to the extracellular matrix, which is necessary to transmit contractility signals between neighbouring cells (reviewed in Sparrow and Schöck, 2009). Thus, Zasp52 appears to act as a key regulator of these signalling dynamics, although it is still unknown if Zasp52 acts directly on integrin or indirectly on Slik, which phosphorylates and thereby regulates talin (Katzemich et al., 2019).

In addition to being involved in adhesion sites, Zasp52 is also a core regulator of Z-disc structure, which delineates the sarcomere and anchors the actin thin filaments in the contractile apparatus. In Drosophila IFMs, Zasp52 localizes to Z-discs during early development and is necessary for the maintenance of the myofibril (Katzemich et al., 2013). In addition to Zasp52, there are two other ALP/Enigma proteins that were discovered in Drosophila, Zasp66 and Zasp67, which are also necessary for the assembly of myofibrils (González-Morales et al., 2019a). Double mutant knockdowns of Zasp52 with either Zasp66 or Zasp67 causes a more severe phenotype than Zasp52 alone or than the α-actinin null mutant phenotype, which is characterized by loss of Z-disc structure (Katzemich et al., 2013). This research suggests that each member of the ALP/Enigma family in Drosophila forms a multi-protein complex with α-actinin that is crucial for Z-disc formation during development. Recently, Zasp52 has also been shown to bind actin filaments directly (Liao et al., 2020). This may explain the incomplete rescue observed with a Zasp52 transgene containing only the α-actinin binding domain. By being able to bind to actin directly, Zasp52 would still be able to anchor Z-discs to the cytoskeleton and retain the structure of the contractile apparatus in some cases of muscle diseases. The region required for actin binding in Zasp52 includes the extended PDZ domain, and the ZM region that immediately follows, although it is still unknown if amino acids of the 26 amino acid ZM domain itself contribute to actin binding (Liao et al., 2020).

Apart from the PDZ and LIM domains, Zasp52 also contains the muscle-specific ZM domain. Recently, using the Drosophila IFM as a model, the importance of this domain has been elucidated (Figure 2B). One common property of sarcomeres is that in all organisms, they have an invariant length and width within one muscle type, and in vertebrates, even show invariance within the species. Until now, the mechanisms behind the determination of said width had not yet been established. The ZM domain may be a key factor in determining width of the sarcomeres. During muscle development, small structures called Z-bodies develop, and are the precursors of the mature sarcomere. These Z-bodies are complexes of α-actinin and associated proteins that grow in size and eventually dictate the final size of the sarcomere (Wang et al., 2005; González-Morales et al., 2019b). As mentioned earlier, Zasp isoforms make part of the Z-disc protein complex, and are critical for proper Z-disc development (Katzemich et al., 2013). All Zasp isoforms in Drosophila have a PDZ and ZM domain, while only Zasp52 contains additional LIM domains. Interestingly, temporal expression and localization of specific isoforms is what determines sarcomere growth. Zasp52, which contains LIM domains, has been shown to be a “growing” isoform of the Z-disc, while Zasp66 and Zasp67, lacking LIM domains, are “blocking isoforms.” The LIM domains and ZM domains interact, and it is this interaction that determines growth of the Z-disc. During the growth phase of the sarcomere, Zasp proteins with unbound LIM domains recruit additional Zasp proteins by the ZM domain. Each of the four LIM domains are able to interact with a ZM domain, which can increase the size of the sarcomere by recruiting additional growing isoforms. However, in the late phase, the ratio of growing to blocking isoforms changes, and more blocking isoforms lacking LIM domains begin binding to the already aggregated Zasp52 molecules. This terminates the growth of the sarcomere by impeding further growing isoform recruitment (González-Morales et al., 2019b).

Having shown the importance of Zasp in muscle cells and the sarcomere, we will now move on to what these discoveries mean for human myopathies, as well as other potential roles that ALP/Enigma proteins play in muscle-related diseases.

Discussion

The pathology of myofibrillar myopathies originates in the Z-disc. Often in myofibrillar myopathies, symptom onset is caused by disintegration of the Z-disc, then the myofibrils, followed by an abnormal ectopic accumulation of multiple proteins (Selcen and Engel 2005). In Zasp-related myopathies, sometimes referred to as zaspopathies, prognosis follows a similar trajectory.

In contrast to Drosophila muscle, vertebrate skeletal muscle has high regenerative properties due to the presence of satellite cells, a group of quiescent muscle progenitors that proliferate, differentiate and fuse into pre-existing myofibers upon muscle injury (reviewed in Kang and Krauss 2010). These newly formed myofibers adopt the stereotypical sarcomeric structure, and undergo a similar molecular mechanism found in developmental myogenesis to reach their final state (reviewed in Bentzinger et al., 2012). This process requires a mechanism to set the diameter of the Z-disc, and therefore it is plausible that the process outlined in González-Morales et al. (2019b) is found in human myogenesis throughout the entire life cycle including muscle development and muscle regeneration caused by muscle damage and during hypertrophy. Interestingly, some Cypher (ZASP) and ENH (PDLIM5) single and double mutants show a similar phenotype than the one found in Drosophila sarcomeres in which the “blocking” isoform predominates, namely a smaller Z-disc diameter (Zhou et al., 2001; Cheng et al., 2010; Mu et al., 2015). In humans, there are ZASP isoforms that lack all LIM domains and are comprised solely of the PDZ and ZM domains, as well as the ALP subfamily in which the proteins only possess one LIM domain (Cheng et al., 2011; reviewed in Zheng et al., 2009). These proteins could represent the “blocking” isoforms in the Drosophila Z-disc diameter model which would function to end Z-disc growth at the specified diameter. Interestingly, in mouse models, when the larger form of ZASP containing all three LIM domains is knocked out, neonatal lethality and Z-disc perturbations are observed, while knock-out of the shorter ZASP isoform containing no LIM domains conversely has no phenotype (Cheng et al., 2011). Moreover, ENH shows a similar pattern of isoform transition from a LIM-containing to LIM-less splice variant during mouse heart development (Yamazaki et al., 2010; Ito et al., 2012). In addition, there are many ZM domain as well as LIM domain mutations found in zaspopathies (Vatta et al., 2003; Selcen and Engel 2005; Theis et al., 2006) which lends further credence to this hypothesis.

Recently, SNPs in PDLIM3 and PDLIM5 have been implicated in the development of idiopathic dilated cardiomyopathy (Wang et al., 2019). Additionally, loss of function variants in PDLIM3 have been correlated with atrial fibrillation, a common cardiac arrhythmia (Vad et al., 2020), as well as hypertrophic cardiomyopathy (Lopes et al., 2015). These recent findings highlight the growing relevance of ALP/Enigma proteins during muscle and heart development, and possible implications for human disease. In mammals, the ALP/Enigma proteins have been proposed to have redundant roles because of their shared domains (Jo et al., 2001; Mu et al., 2015; reviewed in Zheng et al., 2009). However, if mutations are compounded, the redundancy of the proteins may not suffice for proper muscle function. Fichna et al. (2018) have proposed different possible inheritance modes for myofibrillar myopathies in their review, two of which involve the accumulation of minor or benign variants which can in conjunction compound and lead to the development of disease. There is already some evidence to support this with the ALP/Enigma family. Recently, PDLIM5 has been suggested to be a disease modifier in familial DCM cases caused by mutations in the TTN gene (Verdonschot et al., 2019). Additionally, lower mRNA expression of PDLIM5 was shown to be matched with an increase in mRNA expression of ZASP, one of its interactors in the Z-disc and member of the ALP/Enigma family, which validates previous PDLIM5-knockout mouse model data (Cheng et al., 2010). Therefore, it is entirely possible that this process could be relevant for ALP/Enigma proteins in a muscle context. A patient with a benign or minor variation in ZASP may be more susceptible to developing a myopathy if there are other mutations in redundant ALP/Enigma family proteins, such as PDLIM3 or PDLIM5, which are now no longer able to provide the structural integrity of the Z-disc.

The ALP/Enigma proteins are a diverse group of adaptor proteins involved in numerous and vastly different cellular processes. In this review, we highlighted the evolution of their protein structure. It is from the complexity of their domains that their versatility is conferred, which enables them to bind a myriad of different proteins. We also covered recent advances in Drosophila, showcasing the potential implications for muscle disorders. The mounting research in recent years on the ALP/Enigma family proteins is uncovering the families’ relevance for muscle-related diseases and possible therapeutic targets in a clinical setting. More research will need to be carried out in order to elucidate therapeutic avenues, as well as examining the possible genetic interactions between multiple members of the family in a myopathic context. Of particular interest should be future studies investigating if sarcomere diameter is controlled similarly in vertebrates as in insects and what this means for human myopathies.

Author contributions

LF: writing of original draft, editing; FS: writing, editing, funding acquisition and project administration.

Funding

This work was supported by operating grant PJT-155995 to FS from the Canadian Institutes of Health Research (https://cihr-irsc.gc.ca) and RGPIN-2016-06793 to FS from the Natural Sciences and Engineering Research Council of Canada (https://www.nserc-crsng.gc.ca).

Conflict of interest

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

Publisher’s note

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

References

Ahn, B. Y., Saldanha-Gama, R. F., Rahn, J. J., Hao, X., Zhang, J., Dang, N. H., et al. (2015). Glioma invasion mediated by the p75 neurotrophin receptor (p75NTR/CD271) requires regulated interaction with PDLIM1. Oncogene 35, 1411–1422. doi:10.1038/onc.2015.199

PubMed Abstract | CrossRef Full Text | Google Scholar

Arimura, T., Hayashi, T., Terada, H., Lee, S. Y., Zhou, Q., Takahashi, M., et al. (2004). A Cypher/ZASP mutation associated with dilated cardiomyopathy alters the binding affinity to protein kinase C. J. Biol. Chem. 279, 6746–6752. doi:10.1074/jbc.M311849200

PubMed Abstract | CrossRef Full Text | Google Scholar

Arimura, T., Inagaki, N., Hayashi, T., Shichi, D., Sato, A., Hinohara, K., et al. (2009). Impaired binding of ZASP/Cypher with phosphoglucomutase 1 is associated with dilated cardiomyopathy. Cardiovasc. Res. 83, 80–88. doi:10.1093/cvr/cvp119

PubMed Abstract | CrossRef Full Text | Google Scholar

Au, Y., Atkinson, R. A., Guerrini, R., Kelly, G., Joseph, C., Martin, S. R., et al. (2004). Solution structure of ZASP PDZ domain; implications for sarcomere ultrastructure and enigma family redundancy. Structure 12, 611–622. doi:10.1016/j.str.2004.02.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Bang, C., Batkai, S., Dangwal, S., Gupta, S. K., Foinquinos, A., Holzmann, A., et al. (2014). Cardiac fibroblast–derived microRNA passenger strand-enriched exosomes mediate cardiomyocyte hypertrophy. J. Clin. Invest. 124, 2136–2146. doi:10.1172/JCI70577

PubMed Abstract | CrossRef Full Text | Google Scholar

Barrès, R., Gonzalez, T., Marchand-Brustel, Y. L., and Tanti, J.-F. (2005). The interaction between the adaptor protein APS and Enigma is involved in actin organisation. Exp. Cell Res. 308, 334–344. doi:10.1016/j.yexcr.2005.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Barrès, R., Grémeaux, T., Gual, P., Gonzalez, T., Gugenheim, J., Tran, A., et al. (2006). Enigma interacts with adaptor protein with PH and SH2 domains to control insulin-induced actin cytoskeleton remodeling and glucose transporter 4 translocation. Mol. Endocrinol. 20, 2864–2875. doi:10.1210/me.2005-0455

PubMed Abstract | CrossRef Full Text | Google Scholar

Bauer, K., Kratzer, M., Otte, M., de Quintana, K. L., Hagmann, J., Arnold, G. J., et al. (2000). Human CLP36, a PDZ-domain and LIM-domain protein, binds to α-actinin-1 and associates with actin filaments and stress fibers in activated platelets and endothelial cells. Blood 96, 4236–4245. doi:10.1182/blood.v96.13.4236.h8004236_4236_4245

PubMed Abstract | CrossRef Full Text | Google Scholar

Baumert, R., Ji, H., Paulucci-Holthauzen, A., Wolfe, A., Sagum, C., Hodgson, L., et al. (2020). Novel phospho-switch function of delta-catenin in dendrite development. J. Cell Biol. 219, e201909166. doi:10.1083/jcb.201909166

PubMed Abstract | CrossRef Full Text | Google Scholar

Bentzinger, C. F., Wang, Y. X., and Rudnicki, M. A. (2012). Building muscle: Molecular regulation of myogenesis. Cold Spring Harb. Perspect. Biol. 4, a008342. doi:10.1101/cshperspect.a008342

PubMed Abstract | CrossRef Full Text | Google Scholar

Borrello, M. G., Mercalli, E., Perego, C., Degl'Innocenti, D., Ghizzoni, S., Arighi, E., et al. (2002). Differential interaction of Enigma protein with the two RET isoforms. Biochem. Biophys. Res. Commun. 296, 515–522. doi:10.1016/s0006-291x(02)00886-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Bouaouina, M., Jani, K., Long, J. Y., Czerniecki, S., Morse, E. M., Ellis, S. J., et al. (2012). Zasp regulates integrin activation. J. Cell Sci. 125, 5647–5657. doi:10.1242/jcs.103291

PubMed Abstract | CrossRef Full Text | Google Scholar

Bowe, R. A., Cox, O. T., Ayllón, V., Tresse, E., Healy, N. C., Edmunds, S. J., et al. (2014). PDLIM2 regulates transcription factor activity in epithelial-to-mesenchymal transition via the COP9 signalosome. Mol. Biol. Cell 25, 184–195. doi:10.1091/mbc.E13-06-0306

PubMed Abstract | CrossRef Full Text | Google Scholar

Bozulic, L., Malik, M., Powell, D., Nanez, A., Link, A., Ramos, K., et al. (2007). Plasma membrane Ca2+-ATPase associates with CLP36, α-actinin and actin in human platelets. Thromb. Haemost. 97, 587–597. doi:10.1160/th06-08-0438

PubMed Abstract | CrossRef Full Text | Google Scholar

Camarata, T., Krcmery, J., Snyder, D., Park, S., Topczewski, J., and Simon, H. G. (2010). PDLIM7 (LMP4) regulation of TBX5 specifies zebrafish heart atrio-ventricular boundary and valve formation. Dev. Biol. 337, 233–245. doi:10.1016/j.ydbio.2009.10.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Chechenova, M. B., Bryantsev, A. L., and Cripps, R. M. (2013). The Drosophila Z-disc protein Z(210) is an adult muscle isoform of Zasp52, which is required for normal myofibril organization in indirect flight muscles. J. Biol. Chem. 288, 3718–3726. doi:10.1074/jbc.M112.401794

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, H. N., Yuan, K., Xie, N., Wang, K., Huang, Z., Chen, Y., et al. (2015). PDLIM1 stabilizes the E-cadherin/β-catenin complex to prevent epithelial–mesenchymal transition and metastatic potential of colorectal cancer cells. Cancer Res. 76, 1122–1134. doi:10.1158/0008-5472.CAN-15-1962

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Lai, M., Maeno-Hikichi, Y., and Zhang, J. (2006). Essential role of the LIM domain in the formation of the PKCɛ–ENH–N-type Ca2+ channel complex. Cell. Signal. 18, 215–224. doi:10.1016/j.cellsig.2005.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, H., Kimura, K., Peter, A. K., Cui, L., Ouyang, K., Shen, T., et al. (2010). Loss of Enigma homolog protein results in dilated cardiomyopathy. Circ. Res. 107, 348–356. doi:10.1161/CIRCRESAHA.110.218735

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheng, H., Zheng, M., Peter, A. K., Kimura, K., Li, X., Ouyang, K., et al. (2011). Selective deletion of long but not short Cypher isoforms leads to late-onset dilated cardiomyopathy. Hum. Mol. Genet. 20, 1751–1762. doi:10.1093/hmg/ddr050

PubMed Abstract | CrossRef Full Text | Google Scholar

Cuppen, E., Gerrits, H., Pepers, B., Wieringa, B., and Hendriks, W. (1998). PDZ motifs in PTP-BL and RIL bind to internal protein segments in the LIM domain protein RIL. Mol. Biol. Cell 9, 671–683. doi:10.1091/mbc.9.3.671

PubMed Abstract | CrossRef Full Text | Google Scholar

Cuppen, E., van Ham, M., Wansink, D. G., de Leeuw, A., Wieringa, B., and Hendriks, W. (2000). The zyxin-related protein TRIP6 interacts with PDZ motifs in the adaptor protein RIL and the protein tyrosine phosphatase PTP-BL. Eur. J. Cell Biol. 79, 283–293. doi:10.1078/S0171-9335(04)70031-X

PubMed Abstract | CrossRef Full Text | Google Scholar

D'Cruz, R., Plant, P. J., Pablo, L. A., Lin, S., Chackowicz, J., Correa, J., et al. (2016). PDLIM7 is a novel target of the ubiquitin ligase NEDD4-1 in skeletal muscle. Biochem. J. 473, 267–276. doi:10.1042/BJ20150222

PubMed Abstract | CrossRef Full Text | Google Scholar

Durick, K., Gill, G. N., and Taylor, S. S. (1998). SHC and Enigma are both required for mitogenic signaling by RET/PTC2. Mol. Cell. Biol. 18, 2298–2308. doi:10.1128/mcb.18.4.2298

PubMed Abstract | CrossRef Full Text | Google Scholar

Durick, K., Wu, R. Y., Gill, G. N., and Taylor, S. S. (1996). Mitogenic signaling by RET/PTC2 requires association with Enigma via a LIM domain. J. Biol. Chem. 271, 12691–12694. doi:10.1074/jbc.271.22.12691

PubMed Abstract | CrossRef Full Text | Google Scholar

Elbediwy, A., Vanyai, H., Diaz-de-la-Loza, M. D., Frith, D., Snijders, A. P., and Thompson, B. J. (2018). Enigma proteins regulate YAP mechanotransduction. J. Cell Sci. 131, jcs221788. doi:10.1242/jcs.221788

PubMed Abstract | CrossRef Full Text | Google Scholar

Ernst, A., Appleton, B. A., Ivarsson, Y., Zhang, Y., Gfeller, D., Wiesmann, C., et al. (2014). A structural portrait of the PDZ domain family. J. Mol. Biol. 426, 3509–3519. doi:10.1016/j.jmb.2014.08.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Ernst, A., Gfeller, D., Kan, Z., Seshagiri, S., Kim, P. M., Bader, G. D., et al. (2010). Coevolution of PDZ domain–ligand interactions analyzed by high-throughput phage display and deep sequencing. Mol. Biosyst. 6, 1782–1790. doi:10.1039/c0mb00061b

PubMed Abstract | CrossRef Full Text | Google Scholar

Ernst, A., Sazinsky, S. L., Hui, S., Currell, B., Dharsee, M., Seshagiri, S., et al. (2009). Rapid evolution of functional complexity in a domain family. Sci. Signal. 2, ra50. doi:10.1126/scisignal.2000416

PubMed Abstract | CrossRef Full Text | Google Scholar

Fichna, J. P., Maruszak, A., and Żekanowski, C. (2018). Myofibrillar myopathy in the genomic context. J. Appl. Genet. 59, 431–439. doi:10.1007/s13353-018-0463-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Frey, N., and Olson, E. N. (2002). Calsarcin-3, a novel skeletal muscle-specific member of the Calsarcin family interacts with multiple Z-disc proteins. J. Biol. Chem. 277, 13998–14004. doi:10.1074/jbc.M200712200

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, C., Li, Q., Zou, J., Xing, C., Luo, M., Yin, B., et al. (2019). JMJD3 regulates CD4 T cell trafficking by targeting actin cytoskeleton regulatory gene Pdlim4. J. Clin. Invest. 129, 4745–4757. doi:10.1172/JCI128293

PubMed Abstract | CrossRef Full Text | Google Scholar

Fu, J., Yan, P., Li, S., Qu, Z., and Xiao, G. (2010). Molecular determinants of PDLIM2 in suppressing HTLV-I tax-mediated tumorigenesis. Oncogene 29, 6499–6507. doi:10.1038/onc.2010.374

PubMed Abstract | CrossRef Full Text | Google Scholar

Gallardo, R., Ivarsson, Y., Schymkowitz, J., Rousseau, F., and Zimmermann, P. (2010). Structural diversity of PDZ-lipid interactions. ChemBioChem 11, 456–467. doi:10.1002/cbic.200900616

PubMed Abstract | CrossRef Full Text | Google Scholar

González-Morales, N., Marsh, T. W., Katzemich, A., Marescal, O., Xiao, Y. S., and Schöck, F. (2019a). Different evolutionary trajectories of two insect-specific paralogous proteins involved in stabilizing muscle myofibrils. Genetics 212, 743–755. doi:10.1534/genetics.119.302217

PubMed Abstract | CrossRef Full Text | Google Scholar

González-Morales, N., Xiao, Y. S., Schilling, M. A., Marescal, O., Liao, K. A., and Schöck, F. (2019b). Myofibril diameter is set by a finely tuned mechanism of protein oligomerization in Drosophila. eLife 8, e50496. doi:10.7554/eLife.50496

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, Z. S., and Qu, Z. (2021). PDLIM2: Signaling pathways and functions in cancer suppression and host immunity. Biochim. Biophys. Acta. Rev. Cancer 1876, 188630. doi:10.1016/j.bbcan.2021.188630

PubMed Abstract | CrossRef Full Text | Google Scholar

Gupta, S., Braun, A., Morowski, M., Premsler, T., Bender, M., Nagy, Z., et al. (2012). CLP36 is a negative regulator of glycoprotein VI signaling in platelets. Circ. Res. 111, 1410–1420. doi:10.1161/CIRCRESAHA.112.264754

PubMed Abstract | CrossRef Full Text | Google Scholar

Guy, P. M., Kenny, D. A., and Gill, G. N. (1999). The PDZ domain of the LIM protein Enigma binds to β-tropomyosin. Mol. Biol. Cell 10, 1973–1984. doi:10.1091/mbc.10.6.1973

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, H. F., and Beckerle, M. C. (2009). The ALP-Enigma protein ALP-1 functions in actin filament organization to promote muscle structural integrity in Caenorhabditis elegans. Mol. Biol. Cell 20, 2361–2370. doi:10.1091/mbc.e08-06-0584

PubMed Abstract | CrossRef Full Text | Google Scholar

Harris, B. Z., and Lim, W. A. (2001). Mechanism and role of PDZ domains in signaling complex assembly. J. Cell Sci. 114, 3219–3231. doi:10.1242/jcs.114.18.3219

PubMed Abstract | CrossRef Full Text | Google Scholar

Hasegawa, T., Ohno, K., Funahashi, S., Miyazaki, K., Nagano, A., and Sato, K. (2010). CLP36 interacts with Palladin in dorsal root ganglion neurons. Neurosci. Lett. 476, 53–57. doi:10.1016/j.neulet.2010.03.081

PubMed Abstract | CrossRef Full Text | Google Scholar

He, H., Yin, H., Yu, X., Zhang, Y., Ma, M., Li, D., et al. (2021). PDLIM5 affects chicken skeletal muscle satellite cell proliferation and differentiation via the p38-MAPK pathway. Animals. 11, 1016. doi:10.3390/ani11041016

PubMed Abstract | CrossRef Full Text | Google Scholar

Henderson, J. R., Pomiès, P., Auffray, C., and Beckerle, M. C. (2003). Alp and MLP distribution during myofibrillogenesis in cultured cardiomyocytes. Cell Motil. Cytoskelet. 54, 254–265. doi:10.1002/cm.10102

PubMed Abstract | CrossRef Full Text | Google Scholar

Herrick, S., Evers, D. M., Lee, J. Y., Udagawa, N., and Pak, D. T. S. (2010). Postsynaptic PDLIM5/Enigma homolog binds SPAR and causes dendritic spine shrinkage. Mol. Cell. Neurosci. 43, 188–200. doi:10.1016/j.mcn.2009.10.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Horiuchi, Y., Ishikawa, M., Kaito, N., Iijima, Y., Tanabe, Y., Ishiguro, H., et al. (2013). Experimental evidence for the involvement of PDLIM5 in mood disorders in hetero knockout mice. PLoS ONE 8, e59320. doi:10.1371/journal.pone.0059320

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, C., Zhou, Q., Liang, P., Hollander, M. S., Sheikh, F., Li, X., et al. (2003). Characterization and in vivo functional analysis of splice variants of cypher. J. Biol. Chem. 278, 7360–7365. doi:10.1074/jbc.M211875200

PubMed Abstract | CrossRef Full Text | Google Scholar

Huang, J., Cai, C., Zheng, T., Wu, X., Wang, D., Zhang, K., et al. (2020). Endothelial scaffolding protein ENH (Enigma homolog protein) promotes PHLPP2 (pleckstrin homology domain and leucine-rich repeat protein phosphatase 2)-mediated dephosphorylation of AKT1 and eNOS (endothelial NO synthase) promoting vascular remodeling. Arterioscler. Thromb. Vasc. Biol. 40, 1705–1721. doi:10.1161/ATVBAHA.120.314172

PubMed Abstract | CrossRef Full Text | Google Scholar

Iida, Y., Matsuzaki, T., Morishima, T., Sasano, H., Asai, K., Sobue, K., et al. (2009). Localization of reversion-induced Lim protein (RIL) in the rat central nervous system. Acta histochem. cytochem. 42, 9–14. doi:10.1267/ahc.08038

PubMed Abstract | CrossRef Full Text | Google Scholar

Ito, J., Hashimoto, T., Nakamura, S., Aita, Y., Yamazaki, T., Schlegel, W., et al. (2012). Splicing transitions of the anchoring protein ENH during striated muscle development. Biochem. Biophys. Res. Commun. 421, 232–238. doi:10.1016/j.bbrc.2012.03.142

PubMed Abstract | CrossRef Full Text | Google Scholar

Ito, J., Iijima, M., Yoshimoto, N., Niimi, T., Kuroda, S., and Maturana, A. D. (2015). Scaffold protein Enigma homolog activates CREB whereas a short splice variant prevents CREB activation in cardiomyocytes. Cell. Signal. 27, 2425–2433. doi:10.1016/j.cellsig.2015.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Jani, K., and Schöck, F. (2007). Zasp is required for the assembly of functional integrin adhesion sites. J. Cell Biol. 179, 1583–1597. doi:10.1083/jcb.200707045

PubMed Abstract | CrossRef Full Text | Google Scholar

Jia, Y., Shi, H., Cao, Y., Feng, W., Li, M., and Li, X. (2019). PDZ and LIM domain protein 4 suppresses the growth and invasion of ovarian cancer cells via inactivation of STAT3 signaling. Life Sci. 233, 116715. doi:10.1016/j.lfs.2019.116715

PubMed Abstract | CrossRef Full Text | Google Scholar

Jo, K., Rutten, B., Bunn, R. C., and Bredt, D. S. (2001). Actinin-associated Lim protein-deficient mice maintain normal development and structure of skeletal muscle. Mol. Cell. Biol. 21, 1682–1687. doi:10.1128/MCB.21.5.1682-1687.2001

PubMed Abstract | CrossRef Full Text | Google Scholar

Jodo, A., Shibazaki, A., Onuma, A., Kaisho, T., and Tanaka, T. (2020). PDLIM7 synergizes with PDLIM2 and P62/SQSTM1 to inhibit inflammatory signaling by promoting degradation of the p65 subunit of NF-ΚB. Front. Immunol. 11, 1559. doi:10.3389/fimmu.2020.01559

PubMed Abstract | CrossRef Full Text | Google Scholar

Joyce, M. A., Berry-Wynne, K. M., dos Santos, T., Addison, W. R., McFarlane, N., Hobman, T., et al. (2019). HCV and flaviviruses hijack cellular mechanisms for nuclear STAT2 degradation: Up-regulation of PDLIM2 suppresses the innate immune response. PLoS Pathog. 15, e1007949. doi:10.1371/journal.ppat.1007949

PubMed Abstract | CrossRef Full Text | Google Scholar

Jung, C. R., Lim, J. H., Choi, Y., Kim, D. G., Kang, K. J., Noh, S. M., et al. (2010). Enigma negatively regulates p53 through MDM2 and promotes tumor cell survival in mice. J. Clin. Invest. 120, 4493–4506. doi:10.1172/JCI42674

PubMed Abstract | CrossRef Full Text | Google Scholar

Kadrmas, J. L., and Beckerle, M. C. (2004). The LIM domain: From the cytoskeleton to the nucleus. Nat. Rev. Mol. Cell Biol. 5, 920–931. doi:10.1038/nrm1499

PubMed Abstract | CrossRef Full Text | Google Scholar

Kales, S. C., Nau, M. M., Merchant, A. S., and Lipkowitz, S. (2014). Enigma prevents CBL-C-mediated ubiquitination and degradation of RETMEN2A. PLoS ONE 9, e87116. doi:10.1371/journal.pone.0087116

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalyoncu, S., Keskin, O., and Gursoy, A. (2010). Interaction prediction and classification of PDZ domains. BMC Bioinforma. 11, 357. doi:10.1186/1471-2105-11-357

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, J. S., and Krauss, R. S. (2010). Muscle Stem Cells in developmental and regenerative myogenesis. Curr. Opin. Clin. Nutr. Metab. Care 13, 243–248. doi:10.1097/MCO.0b013e328336ea98

PubMed Abstract | CrossRef Full Text | Google Scholar

Katkoori, V. R., Shanmugam, C., Jia, X., Vitta, S. P., Sthanam, M., Callens, T., et al. (2012). Prognostic significance and gene expression profiles of P53 mutations in microsatellite-stable stage III colorectal adenocarcinomas. PLoS ONE 7, e30020. doi:10.1371/journal.pone.0030020

PubMed Abstract | CrossRef Full Text | Google Scholar

Kato, T., Iwayama, Y., Kakiuchi, C., Iwamoto, K., Yamada, K., Minabe, Y., et al. (2005). Gene expression and association analyses of LIM (PDLIM5) in bipolar disorder and schizophrenia. Mol. Psychiatry 10, 1045–1055. doi:10.1038/sj.mp.4001719

PubMed Abstract | CrossRef Full Text | Google Scholar

Katzemich, A., Liao, K. A., Czerniecki, S., and Schöck, F. (2013). ALP/Enigma family proteins cooperate in Z-disc formation and Myofibril Assembly. PLoS Genet. 9, e1003342. doi:10.1371/journal.pgen.1003342

PubMed Abstract | CrossRef Full Text | Google Scholar

Katzemich, A., Long, J. Y., Panneton, V., Fisher, L. A. B., Hipfner, D., and Schöck, F. (2019). Slik phosphorylation of talin T152 is crucial for proper talin recruitment and maintenance of muscle attachment in Drosophila. Development 146, dev176339. doi:10.1242/dev.176339

PubMed Abstract | CrossRef Full Text | Google Scholar

Kimura, A., Kitajima, M., Nishida, K., Serada, S., Fujimoto, M., Naka, T., et al. (2018). NQO1 inhibits the TLR-dependent production of selective cytokines by promoting IκB-ζ degradation. J. Exp. Med. 215, 2197–2209. doi:10.1084/jem.20172024

PubMed Abstract | CrossRef Full Text | Google Scholar

Klaavuniemi, T., Alho, N., Hotulainen, P., Kelloniemi, A., Havukainen, H., Permi, P., et al. (2009). Characterization of the interaction between actinin-associated LIM protein (ALP) and the rod domain of α-actinin. BMC Cell Biol. 10, 22. doi:10.1186/1471-2121-10-22

PubMed Abstract | CrossRef Full Text | Google Scholar

Klaavuniemi, T., Kelloniemi, A., and Ylänne, J. (2004). The ZASP-like motif in actinin-associated LIM protein is required for interaction with the α-actinin rod and for targeting to the muscle Z-line. J. Biol. Chem. 279, 26402–26410. doi:10.1074/jbc.M401871200

PubMed Abstract | CrossRef Full Text | Google Scholar

Klaavuniemi, T., and Ylänne, J. (2006). Zasp/cypher internal ZM-motif containing fragments are sufficient to co-localize with α-actinin—Analysis of patient mutations. Exp. Cell Res. 312, 1299–1311. doi:10.1016/j.yexcr.2005.12.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Klein, M. E., Dickson, M. A., Antonescu, C., Qin, L. X., Dooley, S. J., Barlas, A., et al. (2018). PDLIM7 and CDH18 regulate the turnover of MDM2 during CDK4/6 inhibitor therapy-induced senescence. Oncogene 37, 5066–5078. doi:10.1038/s41388-018-0332-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Koch, B. J., Ryan, J. F., and Baxevanis, A. D. (2012). The diversification of the LIM superclass at the base of the metazoa increased subcellular complexity and promoted multicellular specialization. PLoS ONE 7, e33261. doi:10.1371/journal.pone.0033261

PubMed Abstract | CrossRef Full Text | Google Scholar

Kotaka, M., Kostin, S., Ngai, S., Chan, K., Lau, Y., Lee, S. M., et al. (2000). Interaction of hCLIM1, an Enigma family protein, with α-actinin 2. J. Cell. Biochem. 78, 558–565. doi:10.1002/1097-4644(20000915)78:4<558::aid-jcb5>3.0.co;2-i

PubMed Abstract | CrossRef Full Text | Google Scholar

Krcmery, J., Camarata, T., Kulisz, A., and Simon, H. G. (2010). Nucleocytoplasmic functions of the PDZ-LIM protein family: New insights into organ development. Bioessays 32, 100–108. doi:10.1002/bies.200900148

PubMed Abstract | CrossRef Full Text | Google Scholar

Krcmery, J., Gupta, R., Sadleir, R. W., Ahrens, M. J., Misener, S., Kamide, C., et al. (2013). Loss of the cytoskeletal protein Pdlim7 predisposes mice to heart defects and hemostatic dysfunction. PLoS ONE 8, e80809. doi:10.1371/journal.pone.0080809

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuroda, S., Tokunaga, C., Kiyohara, Y., Higuchi, O., Konishi, H., Mizuno, K., et al. (1996). Protein-protein interaction of zinc finger LIM domains with protein kinase C. J. Biol. Chem. 271, 31029–31032. doi:10.1074/jbc.271.49.31029

PubMed Abstract | CrossRef Full Text | Google Scholar

Lasorella, A., and Iavarone, A. (2006). The protein ENH is a cytoplasmic sequestration factor for Id2 in normal and tumor cells from the nervous system. Proc. Natl. Acad. Sci. U. S. A. 103, 4976–4981. doi:10.1073/pnas.0600168103

PubMed Abstract | CrossRef Full Text | Google Scholar

Lechuga, S., Alarcón, L., Solano, J., Huerta, M., Lopez-Bayghen, E., and González-Mariscal, L. (2010). Identification of ZASP, a novel protein associated to Zona occludens-2. Exp. Cell Res. 316, 3124–3139. doi:10.1016/j.yexcr.2010.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, H. J., and Zheng, J. J. (2010). PDZ domains and their binding partners: Structure, specificity, and modification. Cell Commun. Signal. 8, 8. doi:10.1186/1478-811X-8-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, J. H., Cheng, R., Vardarajan, B., Lantigua, R., Reyes-Dumeyer, D., Ortmann, W., et al. (2015). Genetic modifiers of age at onset in carriers of the G206A mutation in psen1 with familial Alzheimer disease among Caribbean Hispanics. JAMA Neurol. 72, 1043–1051. doi:10.1001/jamaneurol.2015.1424

PubMed Abstract | CrossRef Full Text | Google Scholar

Lemke, S. B., and Schnorrer, F. (2017). Mechanical forces during muscle development. Mech. Dev. 144, 92–101. doi:10.1016/j.mod.2016.11.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Lenfant, N., Polanowska, J., Bamps, S., Omi, S., Borg, J. P., and Reboul, J. (2010). A genome-wide study of PDZ-domain interactions in C. elegans reveals a high frequency of non-canonical binding. BMC Genomics 11, 671. doi:10.1186/1471-2164-11-671

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C., Tao, R., Qin, W., Zheng, Y., He, G., Shi, Y., et al. (2008). Positive association between PDLIM5 and schizophrenia in the Chinese Han population. Int. J. Neuropsychopharmacol. 11, 27–34. doi:10.1017/S1461145707007687

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Z., Ai, T., Samani, K., Xi, Y., Tzeng, H. P., Xie, M., et al. (2010). A ZASP missense mutation, S196L, leads to cytoskeletal and electrical abnormalities in a mouse model of cardiomyopathy. Circ. Arrhythm. Electrophysiol. 3, 646–656. doi:10.1161/CIRCEP.109.929240

PubMed Abstract | CrossRef Full Text | Google Scholar

Liao, K. A., González-Morales, N., and Schöck, F. (2020). Characterizing the actin-binding ability of Zasp52 and its contribution to myofibril assembly. PLoS ONE 15, e0232137. doi:10.1371/journal.pone.0232137

PubMed Abstract | CrossRef Full Text | Google Scholar

Liao, K. A., González-Morales, N., and Schöck, F. (2016). Zasp52, a core Z-disc protein in Drosophila indirect flight muscles, interacts with α-actinin via an extended PDZ domain. PLoS Genet. 12, e1006400. doi:10.1371/journal.pgen.1006400

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, C., Guo, X., Lange, S., Liu, J., Ouyang, K., Yin, X., et al. (2013). Cypher/Zasp is a novel A-kinase anchoring protein. J. Biol. Chem. 288, 29403–29413. doi:10.1074/jbc.M113.470708

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, X., Ruiz, J., Bajraktari, I., Ohman, R., Banerjee, S., Gribble, K., et al. (2014). Z-disc-associated, alternatively spliced, PDZ motif-containing protein (ZASP) mutations in the actin-binding domain cause disruption of skeletal muscle actin filaments in myofibrillar myopathy. J. Biol. Chem. 289, 13615–13626. doi:10.1074/jbc.M114.550418

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, Z., Navarro, V. P., Kempeinen, K. M., Franco, L. M., Jin, Q., Sugai, J. V., et al. (2010). LMP1 regulates periodontal ligament progenitor cell proliferation and differentiation. Bone 47, 55–64. doi:10.1016/j.bone.2010.03.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, H., Pan, H., Yang, H., Wang, J., Zhang, K., Li, X., et al. (2014). LIM mineralization protein-1 suppresses TNF-α induced intervertebral disc degeneration by maintaining nucleus pulposus extracellular matrix production and inhibiting matrix metalloproteinases expression. J. Orthop. Res. 33, 294–303. doi:10.1002/jor.22732

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, X., Chen, L., Huang, H., Lv, J. M., Chen, M., Qu, F. J., et al. (2017). High expression of PDLIM5 facilitates cell tumorigenesis and migration by maintaining AMPK activation in prostate cancer. Oncotarget 8, 98117–98134. doi:10.18632/oncotarget.20981

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Z., Blattner, S. M., Tu, Y., Tisherman, R., Wang, J. H., Rastaldi, M. P., et al. (2011). Alpha-actinin-4 and CLP36 protein deficiencies contribute to podocyte defects in multiple human glomerulopathies. J. Biol. Chem. 286, 30795–30805. doi:10.1074/jbc.M111.255984

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Z., Zhan, Y., Tu, Y., Chen, K., Liu, Z., and Wu, C. (2014). PDZ and Lim Domain Protein 1(PDLIM1)/CLP36 promotes breast cancer cell migration, invasion and metastasis through interaction with α-actinin. Oncogene 34, 1300–1311. doi:10.1038/onc.2014.64

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopes, L. R., Murphy, C., Syrris, P., Dalageorgou, C., McKenna, W. J., Elliott, P. M., et al. (2015). Use of high-throughput targeted exome-sequencing to screen for copy number variation in hypertrophic cardiomyopathy. Eur. J. Med. Genet. 58, 611–616. doi:10.1016/j.ejmg.2015.10.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Lu, Y., Jiang, J., Si, J., Wu, Q., Tian, F., Jiao, K., et al. (2020). PDLIM5 improves depression-like behavior of prenatal stress offspring rats via methylation in male, but not female. Psychoneuroendocrinology 115, 104629. doi:10.1016/j.psyneuen.2020.104629

PubMed Abstract | CrossRef Full Text | Google Scholar

Luck, K., Charbonnier, S., and Travé, G. (2012). The emerging contribution of sequence context to the specificity of protein interactions mediated by PDZ domains. FEBS Lett. 586, 2648–2661. doi:10.1016/j.febslet.2012.03.056

PubMed Abstract | CrossRef Full Text | Google Scholar

Maeda, M., Asano, E., Ito, D., Ito, S., Hasegawa, Y., Hamaguchi, M., et al. (2009). Characterization of interaction between CLP36 and palladin. FEBS J. 276, 2775–2785. doi:10.1111/j.1742-4658.2009.07001.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Maeno-Hikichi, Y., Chang, S., Matsumura, K., Lai, M., Lin, H., Nakagawa, N., et al. (2003). A PKC epsilon–ENH–channel complex specifically modulates N-type Ca2+ channels. Nat. Neurosci. 6, 468–475. doi:10.1038/nn1041

PubMed Abstract | CrossRef Full Text | Google Scholar

Martinelli, V. C., Kyle, W. B., Kojic, S., Vitulo, N., Li, Z., Belgrano, A., et al. (2014). ZASP interacts with the mechanosensing protein Ankrd2 and p53 in the signalling network of striated muscle. PLoS ONE 9, e92259. doi:10.1371/journal.pone.0092259

PubMed Abstract | CrossRef Full Text | Google Scholar

Maturana, A. D., Nakagawa, N., Yoshimoto, N., Tatematsu, K., Hoshijima, M., Tanizawa, K., et al. (2011). LIM domains regulate protein kinase C activity: A novel molecular function. Cell. Signal. 23, 928–934. doi:10.1016/j.cellsig.2011.01.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Maturana, A. D., Wälchli, S., Iwata, M., Ryser, S., Van Lint, J., Hoshijima, M., et al. (2008). Enigma homolog 1 scaffolds protein kinase D1 to regulate the activity of the cardiac L-type voltage-gated calcium channel. Cardiovasc. Res. 78, 458–465. doi:10.1093/cvr/cvn052

PubMed Abstract | CrossRef Full Text | Google Scholar

Moselhy, H., Eapen, V., Akawi, N. A., Younis, A., Salih, B., Othman, A. R., et al. (2015). Secondary Association of PDLIM5 with paranoid schizophrenia in Emirati patients. Meta Gene 5, 135–139. doi:10.1016/j.mgene.2015.07.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Mu, Y., Jing, R., Peter, A. K., Lange, S., Lin, L., Zhang, J., et al. (2015). Cypher and Enigma homolog protein are essential for cardiac development and embryonic survival. J. Am. Heart Assoc. 4, e001950. doi:10.1161/JAHA.115.001950

PubMed Abstract | CrossRef Full Text | Google Scholar

Naegle, K. M., White, F. M., Lauffenburger, D. A., and Yaffe, M. B. (2012). Robust co-regulation of tyrosine phosphorylation sites on proteins reveals novel protein interactions. Mol. Biosyst. 8, 2771–2782. doi:10.1039/c2mb25200g

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakagawa, N., Hoshijima, M., Oyasu, M., Saito, N., Tanizawa, K., and Kuroda, S. (2000). ENH, containing PDZ and LIM domains, heart/skeletal muscle-specific protein, associates with cytoskeletal proteins through the PDZ domain. Biochem. Biophys. Res. Commun. 272, 505–512. doi:10.1006/bbrc.2000.2787

PubMed Abstract | CrossRef Full Text | Google Scholar

Nakatani, M., Ito, J., Koyama, R., Iijima, M., Yoshimoto, N., Niimi, T., et al. (2016). Scaffold protein Enigma homolog 1 overcomes the repression of myogenesis activation by inhibitor of DNA binding 2. Biochem. Biophys. Res. Commun. 474, 413–420. doi:10.1016/j.bbrc.2016.04.119

PubMed Abstract | CrossRef Full Text | Google Scholar

Niederländer, N., Fayein, N. A., Auffray, C., and Pomiès, P. (2004). Characterization of a new human isoform of the Enigma homolog family specifically expressed in skeletal muscle. Biochem. Biophys. Res. Commun. 325, 1304–1311. doi:10.1016/j.bbrc.2004.10.178

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohno, K., Kato, H., Funahashi, S., Hasegawa, T., and Sato, K. (2009). Characterization of CLP36/elfin/PDLIM1 in the nervous system. J. Neurochem. 111, 790–800. doi:10.1111/j.1471-4159.2009.06370.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ohsawa, N., Koebis, M., Suo, S., Nishino, I., and Ishiura, S. (2011). Alternative splicing of PDLIM3/ALP, for α-actinin-associated LIM protein 3, is aberrant in persons with myotonic dystrophy. Biochem. Biophys. Res. Commun. 409, 64–69. doi:10.1016/j.bbrc.2011.04.106

PubMed Abstract | CrossRef Full Text | Google Scholar

Oldfield, C. J., and Dunker, A. K. (2014). Intrinsically disordered proteins and intrinsically disordered protein regions. Annu. Rev. Biochem. 83, 553–584. doi:10.1146/annurev-biochem-072711-164947

PubMed Abstract | CrossRef Full Text | Google Scholar

Ono, R., Kaisho, T., and Tanaka, T. (2015). PDLIM1 inhibits NF-κB-mediated inflammatory signaling by sequestering the p65 subunit of NF-κB in the cytoplasm. Sci. Rep. 5, 18327. doi:10.1038/srep18327

PubMed Abstract | CrossRef Full Text | Google Scholar

Pashmforoush, M., Pomiès, P., Peterson, K. L., Kubalak, S., Ross, J., Hefti, A., et al. (2001). Adult mice deficient in actinin–associated LIM-domain protein reveal a developmental pathway for right ventricular cardiomyopathy. Nat. Med. 7, 591–597. doi:10.1038/87920

PubMed Abstract | CrossRef Full Text | Google Scholar

Pathak, P., Blech-Hermoni, Y., Subedi, K., Mpamugo, J., Obeng-Nyarko, C., Ohman, R., et al. (2021). Myopathy associated LDB3 mutation causes Z-disc disassembly and protein aggregation through PKCα and TSC2-mTOR downregulation. Commun. Biol. 4, 355. doi:10.1038/s42003-021-01864-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Pomiès, P., Macalma, T., and Beckerle, M. C. (1999). Purification and characterization of an α-actinin-binding PDZ-LIM protein that is up-regulated during muscle differentiation. J. Biol. Chem. 274, 29242–29250. doi:10.1074/jbc.274.41.29242

PubMed Abstract | CrossRef Full Text | Google Scholar

Qu, Z., Fu, J., Ma, H., Zhou, J., Jin, M., Mapara, M. Y., et al. (2012). PDLIM2 restricts Th1 and Th17 differentiation and prevents autoimmune disease. Cell Biosci. 2, 23. doi:10.1186/2045-3701-2-23

PubMed Abstract | CrossRef Full Text | Google Scholar

Qu, Z., Yan, P., Fu, J., Jiang, J., Grusby, M. J., Smithgall, T. E., et al. (2010). DNA methylation–dependent repression of PDZ-LIM domain–containing protein 2 in colon cancer and its role as a potential therapeutic target. Cancer Res. 70, 1766–1772. doi:10.1158/0008-5472.CAN-09-3263

PubMed Abstract | CrossRef Full Text | Google Scholar

Ren, B., Li, X., Zhang, J., Fan, J., Duan, J., and Chen, Y. (2015). PDLIM5 mediates PKCε translocation in PMA-induced growth cone collapse. Cell. Signal. 27, 424–435. doi:10.1016/j.cellsig.2014.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Sangadala, S., Boden, S. D., Metpally, R. P., and Reddy, B. V. (2007). Modeling and analysis of molecular interaction between SMURF1-WW2 domain and various isoforms of LIM mineralization protein. Proteins 68, 690–701. doi:10.1002/prot.21429

PubMed Abstract | CrossRef Full Text | Google Scholar

Sangadala, S., Boden, S. D., Viggeswarapu, M., Liu, Y., and Titus, L. (2006). LIM mineralization protein-1 potentiates bone morphogenetic protein responsiveness via a novel interaction with SMURF1 resulting in decreased ubiquitination of Smads. J. Biol. Chem. 281, 17212–17219. doi:10.1074/jbc.M511013200

PubMed Abstract | CrossRef Full Text | Google Scholar

Sangadala, S., Yoshioka, K., Enyo, Y., Liu, Y., Titus, L., and Boden, S. D. (2013). Characterization of a unique motif in LIM mineralization protein-1 that interacts with Jun Activation-domain-binding protein 1. Mol. Cell. Biochem. 385, 145–157. doi:10.1007/s11010-013-1823-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Schiller, H. B., Friedel, C. C., Boulegue, C., and Fässler, R. (2011). Quantitative proteomics of the integrin adhesome show a myosin II-dependent recruitment of LIM domain proteins. EMBO Rep. 12, 259–266. doi:10.1038/embor.2011.5

PubMed Abstract | CrossRef Full Text | Google Scholar

Schulz, T. W., Nakagawa, T., Licznerski, P., Pawlak, V., Kolleker, A., Rozov, A., et al. (2004). Actin/alpha-actinin-dependent transport of AMPA receptors in dendritic spines: Role of the PDZ-LIM protein RIL. J. Neurosci. 24, 8584–8594. doi:10.1523/JNEUROSCI.2100-04.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Selcen, D., and Engel, A. G. (2005). Mutations in ZASP define a novel form of muscular dystrophy in humans. Ann. Neurol. 57, 269–276. doi:10.1002/ana.20376

PubMed Abstract | CrossRef Full Text | Google Scholar

Seto, J. T., Lek, M., Quinlan, K. G. R., Houweling, P. J., Zheng, X. F., Garton, F., et al. (2011). Deficiency of α-actinin-3 is associated with increased susceptibility to contraction-induced damage and skeletal muscle remodeling. Hum. Mol. Genet. 20, 2914–2927. doi:10.1093/hmg/ddr196

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, P., Shathasivam, T., Ignatchenko, V., Kislinger, T., and Gramolini, A. O. (2011). Identification of an FHL1 protein complex containing ACTN1, ACTN4, and PDLIM1 using affinity purifications and MS-based protein–protein interaction analysis. Mol. Biosyst. 7, 1185–1196. doi:10.1039/c0mb00235f

PubMed Abstract | CrossRef Full Text | Google Scholar

Shi, Y., Wang, X., Xu, Z., He, Y., Guo, C., He, L., et al. (2020). PDLIM5 inhibits STUB1-mediated degradation of SMAD3 and promotes the migration and invasion of lung cancer cells. J. Biol. Chem. 295, 13798–13811. doi:10.1074/jbc.RA120.014976

PubMed Abstract | CrossRef Full Text | Google Scholar

Shimizu, T., Pastuhov, S. I., Hanafusa, H., Matsumoto, K., and Hisamoto, N. (2018). The C. elegans BRCA2-ALP/Enigma complex regulates axon regeneration via a Rho GTPase-Rock-MLC phosphorylation pathway. Cell Rep. 24, 1880–1889. doi:10.1016/j.celrep.2018.07.049

PubMed Abstract | CrossRef Full Text | Google Scholar

Sistani, L., Dunér, F., Udumala, S., Hultenby, K., Uhlen, M., Betsholtz, C., et al. (2011). PDLIM2 is a novel actin-regulating protein of podocyte foot processes. Kidney Int. 80, 1045–1054. doi:10.1038/ki.2011.231

PubMed Abstract | CrossRef Full Text | Google Scholar

Sparrow, J. C., and Schöck, F. (2009). The initial steps of myofibril assembly: Integrins pave the way. Nat. Rev. Mol. Cell Biol. 10, 293–298. doi:10.1038/nrm2634

PubMed Abstract | CrossRef Full Text | Google Scholar

Stein, L., Rothschild, J., Luce, J., Cowell, J. K., Thomas, G., Bogdanova, T. I., et al. (2010). Copy number and gene expression alterations in radiation-induced papillary thyroid carcinoma from Chernobyl pediatric patients. Thyroid 20, 475–487. doi:10.1089/thy.2009.0008

PubMed Abstract | CrossRef Full Text | Google Scholar

Strohbach, C., Kleinman, S., Linkhart, T., Amaar, Y., Chen, S. T., Mohan, S., et al. (2008). Potential involvement of the interaction between insulin-like growth factor binding protein (IGFBP)-6 and LIM mineralization protein (LMP)-1 in regulating osteoblast differentiation. J. Cell. Biochem. 104, 1890–1905. doi:10.1002/jcb.21761

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, Y., Hiemstra, T. F., Yan, Y., Li, J., Karet, H. I., Rosen, L., et al. (2017). PDLIM5 links kidney anion exchanger 1 (kAE1) to ILK and is required for membrane targeting of kAE1. Sci. Rep. 7, 39701. doi:10.1038/srep39701

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, X., Phua, D. Y. Z., Axiotakis, L., Smith, M. A., Blankman, E., Gong, R., et al. (2020). Mechanosensing through direct binding of tensed F-actin by LIM domains. Dev. Cell 55, 468–482. doi:10.1016/j.devcel.2020.09.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanaka, T., Grusby, M. J., and Kaisho, T. (2007). PDLIM2-mediated termination of transcription factor NF-kappaB activation by intranuclear sequestration and degradation of the p65 subunit. Nat. Immunol. 8, 584–591. doi:10.1038/ni1464

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanaka, T., Shibazaki, A., Ono, R., and Kaisho, T. (2014). HSP70 mediates degradation of the p65 subunit of nuclear factor κB to inhibit inflammatory signaling. Sci. Signal. 7, ra119. doi:10.1126/scisignal.2005533

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanaka, T., Yamamoto, Y., Muromoto, R., Ikeda, O., Sekine, Y., Grusby, M. J., et al. (2011). PDLIM2 inhibits T helper 17 cell development and granulomatous inflammation through degradation of STAT3. Sci. Signal. 4, ra85. doi:10.1126/scisignal.2001637

PubMed Abstract | CrossRef Full Text | Google Scholar

te Velthuis, A. J. W., Isogai, T., Gerrits, L., and Bagowski, C. P. (2007). Insights into the molecular evolution of the PDZ/LIM family and identification of a novel conserved protein motif. PLoS ONE 2, e189. doi:10.1371/journal.pone.0000189

PubMed Abstract | CrossRef Full Text | Google Scholar

Teyra, J., Ernst, A., Singer, A., Sicheri, F., and Sidhu, S. S. (2019). Comprehensive analysis of all evolutionary paths between two divergent PDZ domain specificities. Protein Sci. 29, 433–442. doi:10.1002/pro.3759

PubMed Abstract | CrossRef Full Text | Google Scholar

Theis, J. L., Bos, J. M., Bartleson, V. B., Will, M. L., Binder, J., Vatta, M., et al. (2006). Echocardiographic-determined septal morphology in Z-disc hypertrophic cardiomyopathy. Biochem. Biophys. Res. Commun. 351, 896–902. doi:10.1016/j.bbrc.2006.10.119

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, C., Moreau, F., Dieterle, M., Hoffmann, C., Gatti, S., Hofmann, C., et al. (2007). The LIM domains of WLIM1 define a new class of actin bundling modules. J. Biol. Chem. 282, 33599–33608. doi:10.1074/jbc.M703691200

PubMed Abstract | CrossRef Full Text | Google Scholar

Tonikian, R., Zhang, Y., Sazinsky, S. L., Currell, B., Yeh, J. H., Reva, B., et al. (2008). A specificity map for the PDZ domain family. PLoS Biol. 6, e239. doi:10.1371/journal.pbio.0060239

PubMed Abstract | CrossRef Full Text | Google Scholar

Torrado, M., Senatorov, V. V., Trivedi, R., Fariss, R. N., and Tomarev, S. I. (2004). PDLIM2, a novel PDZ–LIM domain protein, interacts with α-actinins and filamin A. Invest. Ophthalmol. Vis. Sci. 45, 3955–3963. doi:10.1167/iovs.04-0721

PubMed Abstract | CrossRef Full Text | Google Scholar

Toto, A., Mattei, A., Jemth, P., and Gianni, S. (2017). Understanding the role of phosphorylation in the binding mechanism of a PDZ domain. Protein Eng. Des. Sel. 30, 1–5. doi:10.1093/protein/gzw055

PubMed Abstract | CrossRef Full Text | Google Scholar

Urban, A. E., Quick, E. O., Miller, K. P., Krcmery, J., and Simon, H. G. (2016). Pdlim7 regulates Arf6-dependent actin dynamics and is required for platelet-mediated thrombosis in mice. PLoS ONE 11, e0164042. doi:10.1371/journal.pone.0164042

PubMed Abstract | CrossRef Full Text | Google Scholar

Vad, O. B., Paludan-Müller, C., Ahlberg, G., Kalstø, S. M., Ghouse, J., Andreasen, L., et al. (2020). Loss-of-function variants in cytoskeletal genes are associated with early-onset atrial fibrillation. J. Clin. Med. 9, 372. doi:10.3390/jcm9020372

PubMed Abstract | CrossRef Full Text | Google Scholar

Vallenius, T., Luukko, K., and Mäkelä, T. P. (2000). CLP36 PDZ-LIM protein associates with nonmuscle α-actinin-1 and α-actinin-4. J. Biol. Chem. 275, 11100–11105. doi:10.1074/jbc.275.15.11100

PubMed Abstract | CrossRef Full Text | Google Scholar

Vallenius, T., and Makelä, T. P. (2002). Clik1: A novel kinase targeted to actin stress fibers by the CLP36 PDZ-LIM protein. J. Cell Sci. 115, 2067–2073. doi:10.1242/jcs.115.10.2067

PubMed Abstract | CrossRef Full Text | Google Scholar

Vallenius, T., Scharm, B., Vesikansa, A., Luukko, K., Schäfer, R., and Mäkelä, T. P. (2004). The PDZ-LIM protein RIL modulates actin stress fiber turnover and enhances the association of α-actinin with F-actin. Exp. Cell Res. 293, 117–128. doi:10.1016/j.yexcr.2003.09.004

PubMed Abstract | CrossRef Full Text | Google Scholar

van den Berk, L. C., van Ham, M. A., te Lindert, M. M., Walma, T., Aelen, J., Vuister, G. W., et al. (2005). The interaction of PTP-BL PDZ domains with RIL: An enigmatic role for the RIL LIM Domain. Mol. Biol. Rep. 31, 203–215. doi:10.1007/s11033-005-1407-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Vanaja, D. K., Grossmann, M. E., Cheville, J. C., Gazi, M. H., Gong, A., Zhang, J. S., et al. (2009). PDLIM4, an actin binding protein, suppresses prostate cancer cell growth. Cancer Invest. 27, 264–272. doi:10.1080/07357900802406319

PubMed Abstract | CrossRef Full Text | Google Scholar

Vatta, M., Mohapatra, B., Jimenez, S., Sanchez, X., Faulkner, G., Perles, Z., et al. (2003). Mutations in Cypher/ZASP in patients with dilated cardiomyopathy and left ventricular non-compaction. J. Am. Coll. Cardiol. 42, 2014–2027. doi:10.1016/j.jacc.2003.10.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Verdonschot, J. A., Robinson, E. L., James, K. N., Mohamed, M. W., Claes, G. R., Casas, K., et al. (2019). Mutations in PDLIM5 are rare in dilated cardiomyopathy but are emerging as potential disease modifiers. Mol. Genet. Genomic Med. 8, e1049. doi:10.1002/mgg3.1049

PubMed Abstract | CrossRef Full Text | Google Scholar

von Nandelstadh, P., Ismail, M., Gardin, C., Suila, H., Zara, I., Belgrano, A., et al. (2009). A class III PDZ binding motif in the myotilin and fatz families binds enigma family proteins: A common link for Z-disc myopathies. Mol. Cell. Biol. 29, 822–834. doi:10.1128/MCB.01454-08

PubMed Abstract | CrossRef Full Text | Google Scholar

Walma, T., Spronk, C. A. E. M., Tessari, M., Aelen, J., Schepens, J., Hendriks, W., et al. (2002). Structure, dynamics and binding characteristics of the second PDZ domain of PTP-BL. J. Mol. Biol. 316, 1101–1110. doi:10.1006/jmbi.2002.5402

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, C. K., Pan, L., Chen, J., and Zhang, M. (2010). Extensions of PDZ domains as important structural and functional elements. Protein Cell 1, 737–751. doi:10.1007/s13238-010-0099-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, D., Fang, J., Lv, J., Pan, Z., Yin, X., Cheng, H., et al. (2019). Novel polymorphisms in PDLIM3 and PDLIM5 gene encoding Z-line proteins increase risk of idiopathic dilated cardiomyopathy. J. Cell. Mol. Med. 23, 7054–7062. doi:10.1111/jcmm.14607

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Shaner, N., Mittal, B., Zhou, Q., Chen, J., Sanger, J. M., et al. (2005). Dynamics of Z-band based proteins in developing skeletal muscle cells. Cell Motil. Cytoskelet. 61, 34–48. doi:10.1002/cm.20063

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, K. S., Liu, X., Zhang, Q., Aragam, N., and Pan, Y. (2012). Parent-of-origin effects of FAS and PDLIM1 in attention-deficit/hyperactivity disorder. J. Psychiatry Neurosci. 37, 46–52. doi:10.1503/jpn.100173

PubMed Abstract | CrossRef Full Text | Google Scholar

Watts, N. R., Zhuang, X., Kaufman, J. D., Palmer, I. W., Dearborn, A. D., Coscia, S., et al. (2017). Expression and purification of ZASP subdomains and clinically important isoforms: High-affinity binding to G-actin. Biochemistry 56, 2061–2070. doi:10.1021/acs.biochem.7b00067

PubMed Abstract | CrossRef Full Text | Google Scholar

Winkelman, J. D., Anderson, C. A., Suarez, C., Kovar, D. R., and Gardel, M. L. (2020). Evolutionarily diverse LIM domain-containing proteins bind stressed actin filaments through a conserved mechanism. Proc. Natl. Acad. Sci. U. S. A. 117, 25532–25542. doi:10.1073/pnas.2004656117

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, R. Y., and Gill, G. N. (1994). LIM domain recognition of a tyrosine-containing tight turn. J. Biol. Chem. 269, 25085–25090. doi:10.1016/s0021-9258(17)31502-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Xia, H., Winokur, S. T., Kuo, W. L., Altherr, M. R., and Bredt, D. S. (1997). Actinin-associated LIM protein: Identification of a domain interaction between PDZ and spectrin-like repeat motifs. J. Cell Biol. 139, 507–515. doi:10.1083/jcb.139.2.507

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamashita, Y., Matsuura, T., Kurosaki, T., Amakusa, Y., Kinoshita, M., Ibi, T., et al. (2014). LDB3 splicing abnormalities are specific to skeletal muscles of patients with myotonic dystrophy type 1 and alter its PKC binding affinity. Neurobiol. Dis. 69, 200–205. doi:10.1016/j.nbd.2014.05.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamazaki, T., Wälchli, S., Fujita, T., Ryser, S., Hoshijima, M., Schlegel, W., et al. (2010). Splice variants of Enigma homolog, differentially expressed during heart development, promote or prevent hypertrophy. Cardiovasc. Res. 86, 374–382. doi:10.1093/cvr/cvq023

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, Y., Tsukamoto, O., Nakano, A., Kato, H., Kioka, H., Ito, N., et al. (2015). Augmented AMPK activity inhibits cell migration by phosphorylating the novel substrate PDLIM5. Nat. Commun. 6, 6137. doi:10.1038/ncomms7137

PubMed Abstract | CrossRef Full Text | Google Scholar

Yi, C. R., Allen, J. E., Russo, B., Lee, S. Y., Heindl, J. E., Baxt, L. A., et al. (2014). Systematic analysis of bacterial effector-postsynaptic density 95/disc large/zonula occludens-1 (PDZ) domain interactions demonstrates Shigella OspE protein promotes protein kinase C activation via PDLIM proteins. J. Biol. Chem. 289, 30101–30113. doi:10.1074/jbc.M114.595868

PubMed Abstract | CrossRef Full Text | Google Scholar

Yin, H., Zhao, J., He, H., Chen, Y., Wang, Y., Li, D., et al. (2020). GGA-Mir-3525 targets PDLIM3 through the MAPK signaling pathway to regulate the proliferation and differentiation of skeletal muscle satellite cells. Int. J. Mol. Sci. 21, 5573. doi:10.3390/ijms21155573

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoo, J. Y., Jung, N. C., Lee, J. H., Choi, S. Y., Choi, H. J., Park, S. Y., et al. (2019). PDLIM4 is essential for CCR7-JNK–mediated dendritic cell migration and F-actin–related dendrite formation. FASEB J. 33, 11035–11044. doi:10.1096/fj.201901031

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, J., Li, X., Wang, Y., Li, B., Li, H., Li, Y., et al. (2011). PDLIM2 selectively interacts with the PDZ binding motif of highly pathogenic avian H5N1 influenza A virus NS1. PLoS ONE 6, e19511. doi:10.1371/journal.pone.0019511

PubMed Abstract | CrossRef Full Text | Google Scholar

Yuan, L., Fairchild, M. J., Perkins, A. D., and Tanentzapf, G. (2010). Analysis of integrin turnover in fly myotendinous junctions. J. Cell Sci. 123, 939–946. doi:10.1242/jcs.063040

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, M., Cheng, H., Banerjee, I., and Chen, J. (2009). ALP/Enigma PDZ-LIM domain proteins in the heart. J. Mol. Cell Biol. 2, 96–102. doi:10.1093/jmcb/mjp038

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, M., Cheng, H., Li, X., Zhang, J., Cui, L., Ouyang, K., et al. (2008). Cardiac-specific ablation of Cypher leads to a severe form of dilated cardiomyopathy with premature death. Hum. Mol. Genet. 18, 701–713. doi:10.1093/hmg/ddn400

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, J. K., Fan, X., Cheng, J., Liu, W., and Peng, Y. (2021). PDLIM1: Structure, function and implication in cancer. Cell Stress 5, 119–127. doi:10.15698/cst2021.08.254

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Q., Chu, P. H., Huang, C., Cheng, C. F., Martone, M. E., Knoll, G., et al. (2001). Ablation of Cypher, a PDZ-LIM domain Z-line protein, causes a severe form of congenital myopathy. J. Cell Biol. 155, 605–612. doi:10.1083/jcb.200107092

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Q., Ruiz-Lozano, P., Martone, M. E., and Chen, J. (1999). Cypher, a striated muscle-restricted PDZ and LIM domain-containing protein, binds to α-actinin-2 and protein kinase C. J. Biol. Chem. 274, 19807–19813. doi:10.1074/jbc.274.28.19807

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: ALP/Enigma, PDLIM, PDZ, LIM, ZM, muscle, sarcomere, ZASP

Citation: Fisher LAB and Schöck F (2022) The unexpected versatility of ALP/Enigma family proteins. Front. Cell Dev. Biol. 10:963608. doi: 10.3389/fcell.2022.963608

Received: 08 June 2022; Accepted: 22 November 2022;
Published: 01 December 2022.

Edited by:

Andrea Erika Münsterberg, University of East Anglia, United Kingdom

Reviewed by:

Hiroshi Qadota, Emory University, United States
Elisabeth Ehler, King’s College London, United Kingdom

Copyright © 2022 Fisher and Schöck. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Frieder Schöck, frieder.schoeck@mcgill.ca

Download