Biol. Chem. 2019; 400(8): 1005–1022

Review

Ulrich Kück*, Daria Radchenko and Ines Teichert STRIPAK, a highly conserved signaling complex, controls multiple eukaryotic cellular and developmental processes and is linked with diseases https://doi.org/10.1515/hsz-2019-0173 networks are now known to underlie the transmission and Received February 27, 2019; accepted March 28, 2019; ­previously modulation of such signals. These networks, controlled ­published online May 1, 2019 by diverse regulators at different cellular levels, are highly conserved across eukaryotic organisms. Abstract: The striatin-interacting phosphatases and Among the signaling components that have received kinases (STRIPAK) complex is evolutionary highly con- increased attention in the last decade is the STRIPAK served and has been structurally and functionally complex, which was initially identified in mammals by described in diverse lower and higher eukaryotes. In recent mass spectrometry (MS) analysis (Goudreault et al., 2009). years, this complex has been biochemically characterized Here we will provide a summary of the key studies leading better and further analyses in different model systems have to the discovery of striatin, the major regulatory phos- shown that it is also involved in numerous cellular and phatase subunit of the STRIPAK complex. Further, the developmental processes in eukaryotic organisms. Fur- components and architecture as well as the assembly and ther recent results have shown that the STRIPAK complex structural insights of the STRIPAK complex will be com- functions as a macromolecular assembly communicating prehensively reviewed. Another focus will be the control through physical interaction with other conserved signal- of developmental processes as a result of bi-directional ing complexes to constitute larger dynamic protein interaction of STRIPAK with other components involved networks. Here, we will provide a comprehensive and up- in conserved signaling pathways. Although we address to-date overview of the architecture, function and regula- all aspects of the current STRIPAK research, we mainly tion of the STRIPAK complex and discuss key issues and focus on results obtained in insect, fungal and mamma- future perspectives, linked with human diseases, which lian systems. For further reading, other review articles may form the basis of further research endeavors in this on the STRIPAK complex should provide any remaining area. In particular, the investigation of bi-directional inter- information (Hwang and Pallas, 2014, Kück et al., 2016, actions between STRIPAK and other signaling pathways Shi et al., 2016). should elucidate upstream regulators and downstream tar- gets as fundamental parts of a complex cellular network.

Keywords: Hippo; kinases; striatin; striatin-interacting phosphatases and kinases complex. Discovery of striatin

Striatin is the eponymous component of the STRIPAK complex, and was first detected as an abundant

Introduction synaptosomal protein of 110 000 Mr. In particular, stri- atin is visible in the dorsal part of the striatum as well as In a changing environment, organisms adapt to a multi- in motor neurons, although it is absent in axons, but is tude of extracellular stimuli and cues. Intracellular signal highly abundant in dendritic spines (Castets et al., 1996; transduction pathways forming interconnected regulatory Goudreault et al., 2009). Dendritic spines are tiny, actin- rich protrusions that extend from dendrites and are the sites of most of the excitatory synapses in the mammalian *Corresponding author: Ulrich Kück, Allgemeine und Molekulare CNS. Further analysis using peptide sequencing identi- Botanik, Ruhr-Universität Bochum, Universitätsstr. 150, D-44780 fied a cDNA coding for a 780-amino acids protein, con- Bochum, Germany, e-mail: [email protected] Daria Radchenko and Ines Teichert: Allgemeine und Molekulare taining a caveolin-binding motif, a coiled-coil structure, 2+ Botanik, Ruhr-Universität Bochum, Universitätsstr. 150, D-44780 a Ca -calmodulin-binding domain, and multiple WD40 Bochum, Germany repeats. Successive analyses in mammals found paralogs

Open Access. © 2019 Ulrich Kück et al., published by De Gruyter. This work is licensed under the Creative Commons Attribution- NonCommercial-NoDerivatives 4.0 License. 1006 U. Kück et al.: STRIPAK in development and disease of striatin, which were termed STRN3 (SG2NA) and STRN4 This biochemical characterization defined the major (Zinedin), while the originally described striatin is referred subunits of the human STRIPAK complex, which are to as STRN1. Of note is that striatin homologs have not only the structural (PP2AA) and catalytic (PP2Ac) subunits been found in brain, but also in other tissues such as liver of PP2A, the B‴ regulatory subunits of PP2A (striatins), and cardiac muscle. As outlined later, striatins are regula- striatin-interacting STRIP1/2, Mob3/phocein, cer- tory subunits of protein phosphatase PP2A (Moreno et al., ebral cavernous malformation 3 (CCM3), the sarcolemmal 2000). These subunits are classified into four protein fam- membrane-associated protein (SLMAP), and the coiled- ilies, B, B′, B″, and B‴, where striatins represent the B‴ coil protein suppressor of IκB kinase-ε (IKKε), designated family (Sents et al., 2012). SIKE (Goudreault et al., 2009). A comprehensive overview In Drosophila melanogaster a striatin homolog was of STRIPAK subunits is provided in Table 1, which further identified in a genetic screen for mutations that affect lists the synonymous designations of homologous subu- the dorsal closure in the fly embryo. There, the striatin nits. In animals in general and in mammals in particular, homolog was named connector of kinase to AP-1 (cka) there are several variants of the STRIPAK complex due to and is known to play a role in the JUN N-terminal kinase the existence of isoforms and paralogs of many STRIPAK (DJNK) signal transduction pathway. Interestingly, CKA subunits. For example, STRN1, -3, and -4 are human stri- homologs from Caenorhabditis elegans and mammals can atin paralogs, and their diversity may reflect the many activate the DJNK signal transduction pathway in D. mela- cellular functions of STRIPAK in diseases of higher eukar- nogaster, indicating that they are structural and func- yotes (see below). Homologs of all these subunits were tional homologs (Chen et al., 2002). found in invertebrates, while diverse eukaryotic microbes In non-animal systems, the striatin homolog was possess only a single striatin protein. first identified in a screen for developmental proteins A recently discovered core component of STRIPAK from the sexually propagating fungus Sordaria macros- is SIKE, a suppressor protein of IKKε, which is not con- pora. This ascomycetous fungus was used to generate a served by sequence similarity between closely related genetic library of about 100 sterile mutants, showing a species (Reschka et al., 2018). First discovered in mam- defect in sexual fruiting body formation. Complementa- malian systems (Goudreault et al., 2009), this protein was tion analysis of mutant pro11 revealed a mutated , identified later in the Schizosaccharomyces pombe later named pro11, which has a truncated open reading (Singh et al., 2011), and more recently, in filamentous frame. The PRO11 protein shows a striatin-typical domain fungi (Reschka et al., 2018, Elramli et al., 2019). To date, structure with a conserved coiled-coil region close to its all STRIPAK-associated small coiled-coil proteins identi- N-terminus, a calmodulin , and a C-terminal fied contain one to four predicted coiled-coil regions. domain with seven WD40 repeats (Pöggeler and Kück, Significant components of the STRIPAK complex are 2004). Most remarkably, the sterile pro11 mutant of S. germinal center kinases (GCK), which in mammalian macrospora regained fertility after transformation with a systems can substantially vary, reflecting the different mouse striatin cDNA. After this early discovery of a func- cellular functions of STRIPAK during eukaryotic develop- tional striatin homolog in S. macrospora, many other ment. STRIPAK kinases are members of the GCK family homologs were identified in a huge variety of fungi (see (Kyriakis, 1999). These are a subdivision of the Ste20-like for a review Kück et al., 2016), and a recent molecular evo- kinases, which were named after the Ste20p kinase from lutionary comparison showed that homologs of striatin . GCKs are much more numer- or other STRIPAK subunits are encoded in genomes of a ous in metazoan kinomes than in yeast and are involved broad variety of microbial eukaryotes. This includes slime in a multitude of essential processes. Being evolutionar- molds, which are believed to belong to the early-emerging ily conserved, GCKs are present in both higher and lower eukaryotes during evolution (Nguyen et al., 2018). eukaryotes. In mammals, eight (I-VIII) GCK subfamilies are known, each containing two to four members (Table 2). Some members of these subfamilies transmit extracellu- lar signals to mitogen-activated protein kinase (MAPK) Components and architecture of cascades, others act as signaling hubs within conserved STRIPAK eukaryotic complexes (Delpire, 2009). Although human GCKs and their counterparts from D. melanogaster have The first evidence for a striatin-containing multipro- been extensively characterized and new GCK targets, tein complex came from affinity purification and MS regulators, and interaction partners are being identified, (AP-MS) studies focused on the human PP2A complex. there are still some gaps in our current understanding of U. Kück et al.: STRIPAK in development and disease 1007

Table 1: STRIPAK subunits in selected eukaryotes.

Subunit Function Hs Dm Nc Sm An Sc Sp

Striatin PP2A regulatory subunit STRN1/3/4a Cka HAM-3 PRO11 StrA Far8b Csc3 PP2AA PP2A scaffolding subunit PP2AAα/β Pp2A-29B PP2A-Ac PP2AA SipF Tpd3 Paa1 PP2Ac PP2A catalytic subunit PP2Acα/β Mts PP2Ac PP2Ac1 SipE Pph21/22, Ppg1 Ppa3 STRIP Cell size control STRIP1/2d CG11526 HAM-2 PRO22 SipC Far11 Csc2 Mob Vesicular trafficking Mob3e Mob4 MOB-3 SmMOB3 SipA – – SLMAP Membrane anchoring SLMAP CG17494 HAM-4 PRO45 SipD Far9/10 Csc1 GCKIII Germinal center kinase Stk24/25, Hpo, Msn, ?f SmKIN3 ? ?f (GCKIII) Mst4 GckIII SmKIN24 Small coiled-coil ? SIKE/ FGOP2 NCU04324 SCI1 SipA Far3/7 Csc4p protein FGFR1OP2 CCM3 Tethering of GCKIIIs CCM3 CG5073 – – – – –

In the S. cerevisiae and S. pombe, STRIPAK homologues are named FAR and SIP complexes, respectively. Table modified from diverse references (Goudreault et al., 2009), (Pracheil et al., 2012), (Frost et al., 2012), (Bloemendal et al., 2012), (Ribeiro et al., 2010) and (Hwang and Pallas, 2014). An, A. nidulans; Dm, D. melanogaster; Hs, H. sapiens; Nc, N. crassa; Sc, S. cerevisiae; Sm, S. macrospora; Sp, S. pombe. aMembers of the striatin familiy are termed STRN1 (striatin), STRN3 (SG2NA), and STRN4 (Zinedin). bFar8 lacks the C-terminal WD propeller domain of other homologues of striatin. cPP2A-A has also been termed PPG-1 (Dettmann et al., 2013; Fu et al., 2011). dSTRIP1/2 have also been known as FAM40A and FAM40B, respectively (e.g. Bai et al., 2011). eAlso named phocein. fThe homologous of STRIPAK kinases in N. crassa and S. cerevisiae are SID-1, MST-1, and Sps1, respectively. However, evidence is still lacking that they are subunits of STRIPAK.

Table 2: Ste20-like kinases in mammals and selected ascomycetes.

Family Subfamily Mammals Ascomycetes

Hs Sp Sc An Nc

Germinal center kinases (GCK) GCK-I MAP4K1/2/3/5 Cdc7p Cdc15p SepH CDC-7 GCK-II MST1/2 GCK-III MST3/4, YSK1 Nak1p Kic1p AnPod6 POD-6 GCK-IV MAP4K4/6, NRK, TNIK GCK-V LOK, SLK Ppk11p Sps1p AnMst1 MST-1 GCK-VI SPAK, OSR1 GCK-VII MYO3A/B Sid1p SepL SID-1 GCK-VIII TAO1/2/3 p21 activated kinases (PAK) PAK-I PAK1/2/3 Shk1p Ste20p AnSte20 STE-20 PAK-II PAK4/5/6 Shk2p Cla4p AnCla4 CLA-4

Hs, H. sapiens; Nc, N. crassa; Sc, S. cerevisiae; Sp, S. pombe. Note, there is no subdivision in subfamilies in fungal Ste20-like kinases. Modified after Delpire, 2009, Boyce and Andrianopoulos, 2011. their mechanism and function. GCKs are known to regu- evolutionary conserved within animal systems, it has not late key developmental processes such as cytoskeleton yet been detected in microbial eukaryotes. organization, cell cycle and . These kinases In all eukaryotes, core subunits of STRIPAK are were intensively investigated in some unicellular and structurally conserved, having characteristic functional filamentous ascomycetes (Table 2) and, for example, a domains as depicted in Figure 1. For example, stri- total of four conserved GCKs were described for S. pombe, atins share four conserved domains, a caveolin-binding which are not divided in further subfamilies. domain, a coiled-coil domain, a calmodulin-binding Of special interest is that the key exception of the domain, and WD40 repeat domains. Overall, all STRIPAK overall conservation of STRIPAK subunits seems to be subunits are characterized by several kinds of domains, CCM3, which was found only in animal systems. CCM3 has some of which mediate their interaction with each other. no known functional domain, but yet appears to have the Thus, the composition of STRIPAK complexes seems to be function of binding GCKs to STRIPAK. Although CCM3 is very similar in diverse eukaryotes. However, insights into 1008 U. Kück et al.: STRIPAK in development and disease

STRIATIN CV CC CM WD40 repeats of STRIPAK was refined in a study that investigated the

PP2Ac PP2Ac assembly of Hippo kinase MST2-containing STRIPAK

PP2AA HEAT repeats complex (Tang et al., 2019). Using human cell cultures, the authors propose that a PP2AA/PP2Ac-bound stri- STRIPs CC & TM potential TMs atin contacts MST2, which then is loaded in a phos- SLMAP FHACCTCC M phorylation-dependent manner to STRIP1 and to a MOB3 MOB1-Phocein SIKE-SLMAP heterodimer. Interestingly, a very similar MST3/4, YSK1 STKc assembly process has recently been proposed for a fungal STRIPAK complex (Elramli et al., 2019). Using Figure 1: Primary structure of subunits of the STRIPAK complex. a diverse set of STRIPAK mutants from the ascomycete The general structure of all subunits is similar in all eukaryotes. Modified in part from Frost et al. (2012). Abbreviations: CC, coiled- Aspergillus nidulans, these investigators predict three coil; CM, calmodulin binding domain; CV, caveolin binding domain; heteromeric sub-complexes, which are (i) MOB3 and stri- FHA, forkhead associated domain; MOB1, monopolar spindle-one- atin, (ii) SIKE and SLMAP, and (iii) PP2Ac, PP2AA and binder (phocein); STKc, catalytic domain of serine threonine kinase; STRIP1/2. As shown in Figure 3, these sub-complexes TM, trans-membrane domain; HEAT, Huntingtin, finally form the functional STRIPAK complex. Common 3 (EF3), protein phosphatase 2A (PP2A), and the yeast kinase TOR1; PP2Ac, catalytic domain of protein phosphatase 2A; WD40, WD or to all the structural models is the prediction that stri- beta- repeat sequence. For references see text. atins act as a central scaffold in the STRIPAK complex, directly binding all other subunits. So far however, the direct or indirect interaction of kinases with striatin has the assembly and structure of the complex itself are just not yet been ultimately solved. In mammalian systems, emerging. adaptor proteins promote the interaction of MST3/4 with Crystal structure and biophysical analyses of striatin striatin (Chen et al., 2014). However, MST1/2 pull-down showed that the protein has a parallel dimeric confor- experiments indicate a direct interaction (Tang et al., mation with a large bend (Chen et al., 2014). The coiled- 2019). Similarly, pull-down as well as yeast-two-hybrid coil domain from striatin forms a stable core complex experiments provided evidence for a direct interaction with PP2AA in a 2:2 stoichiometry. Essential for this of fungal MST1/2-like kinase SmKIN3 and MST3/4-like interaction and thus for the assembly of STRIPAK is the kinase SmKIN24 with striatin (Frey et al., 2015). homodimerization of striatin. In particular, the coiled- coil domain seems to be necessary for the assembly of

MOB3 STRIP STRIPAK as it mediates the oligomerization of striatins. 1/2 SIKE As shown in Figure 2, this minimal core complex is able SLMAP PP2AA PP2Ac to associate with GCKs when assisted by adaptor pro-

Cytoplasm Striatin teins. More recently, this architecture and substructure SIKE SLMAP

MOB3 Striatin

Transcription ERK? factors STRIP PP2AA 1/2

Cell cycle PP2Ac

Other genes???

JNK, p38? JNK, p38? Coiled coil Striatins Nucleus factors Stress genes WD40 Adaptor PP2Ac P GCKs PP2AA Figure 3: Model of the assembly of the STRIPAK complex and its role in fungal development (adopted from Elramli et al., 2019). The STRIPAK complex is assembled from three sub-complexes, Figure 2: Physical interactions of subunits of the animal STRIPAK where striatin acts as a scaffold at the . Striatin complex. prevents the shuttling of the JNK homologous MAPK under non- Adaptor, cerebral cavernous malformation 3 (CCM3), adaptor stress conditons and further promotes the phosphorylation of the connecting striatin with GCKs; GCK, germinal center kinase; all ERK homologous MAPK, which in fungi is part of the pheromone other abbreviations are given in the legend of Figure 1. Modified response pathway (PR). For clarity reasons, only the designations from Chen et al. (2014). from the human STRIPAK complex were used. U. Kück et al.: STRIPAK in development and disease 1009

Cellular localization STRIPAK connects several cellular

To elucidate the function of STRIPAK components, cel- signaling complexes lular localization studies were performed in diverse Eukaryotic developmental processes, such as growth, organisms. A comprehensive overview is provided in movement, cell division and differentiation, are governed Table 3, which summarizes most of these studies report- by intertwined signaling networks. These networks are ing distinct subunits of STRIPAK. For example, localiza- important for maintaining homeostasis as they allow tion of STRIPAK subunits at the nuclear envelope has a suitable response to intra- and extracellular stimuli, been described in mammalian cells, insects, worms, including nutrients, stressors and pheromones. Some and unicellular as well as multicellular fungi (reviewed modules of these networks evolved as paralogs, which can by Hwang and Pallas, 2014). Moreover, studies in fungi partially adopt the function of each other in different path- revealed that localization of STRIPAK to the nuclear ways. Other modules have developed independently but envelope depends on striatin and STRIP subunits (Dett- show functional interaction during biological processes. mann et al., 2013; Nordzieke et al., 2015; Elramli et al., Although considerable efforts have been made to 2019). An exception is the S. cerevisae Far complex, where identify upstream regulators and downstream targets of the SLMAP paralogs Far9p and Far10p are required for the STRIPAK complex, our understanding of the mecha- assembly of the complex (Pracheil and Liu, 2013). These nistic function of the STRIPAK complex is still fragmen- results indicate that the yeast Far complex has adapted a tary. STRIPAK complexes have the capacity to regulate different function from other STRIPAK complexes (Frost phosphorylation of diverse proteins, some of which are et al., 2012). Finally, STRIPAK at the nuclear envelope members of associated signaling complexes. As described has also been shown to control MAPK trafficking, as will below, STRIPAK interacts with conserved signaling path- be described later. ways and mediates remodeling of the cytoskeleton. Single subunits of STRIPAK have also been found at other cellular locations. For example, striatin and STRIP have been detected at the nuclear envelope and the Golgi, while SLMAP homologs reside at mitochon- The yeast STRIPAK homolog dria and the endoplasmic reticulum (Byers et al., 2009; counteracts the TORC2 complex Frost et al., 2012; Nordzieke et al., 2015). Switching of SLMAP between nuclear envelope and mitochondria is In budding yeast S. cerevisiae, the STRIPAK homolog is likely conveyed by different tail anchor domains. These the Far complex, which is involved in pheromone-induced C-terminal transmembrane domains direct co-transla- cell cycle arrest, vacuolar protein sorting, and cell fitness. tional integration into target membranes (Byers et al., Far8p is thought to be the striatin homolog, although it 2009). These different localizations of STRIPAK subu- lacks a large part of the C-terminal WD 40 repeat domains nits led to the hypothesis that STRIPAK bridges differ- of striatins, which are important for the function of stri- ent organelles, i.e. the nuclear envelope, the Golgi, and atin in eukaryotic multicellular development (Kemp and the spindle pole body (SPB) or centrosome (the SPB is Sprague, 2003; Frost et al., 2012; Pracheil et al., 2012). the fungal equivalent to the centrosome). This bridging The Far complex directly affects signaling by the TORC2 function may facilitate communication between orga- complex. TOR (target of rapamycin) is a highly conserved nelles during cell division (Frost et al., 2012). Recently, serine-threonine protein kinase. In yeast, it exists in two D. melanogaster striatin has been located at autophago- structurally and functionally distinct complexes, namely somes, and a function of STRIPAK in autophagosome TORC1 and TORC2. Yeast Tor2 kinase, the main organizer trafficking has been recently proposed (Neisch et al., of TORC2, is an important regulator of multiple cellular 2017). functions, including cell growth or actin polymeriza- In conclusion, a conserved localization of the tion (Bartlett and Kim, 2014). A genetic screen with yeast STRIPAK complex was observed at the nuclear envelope, showed that a mutation of FAR11, the homolog of STRIP1/2, where it likely coordinates conserved cellular and devel- suppresses lethality of TORC2-deficient mutants. Further, opmental processes. However, further investigations are a far11 deletion mutant restores phosphorylation of two required to address the question of how different localiza- substrates of the TORC2 complex. Thus, Far antagonizes tions of single subunits may relate to different functions of TORC2 signaling by promoting dephosphorylation of STRIPAK sub-complexes. TORC2 substrates (Pracheil and Liu, 2013). 1010 U. Kück et al.: STRIPAK in development and disease Reference 2019 et al., Elramli 2010 et al., Wang 2016 et al., Maheshwari 2017 et al., Sabour 2009 et al., Byers 2017 et al., Neisch 2018 et al., Zhang 2012 et al., Frost 2012 et al., Frost 2014 Goswami, and Tanti 2013 et al., Dettmann 2017 Jain et al., Liu, 2013 and Pracheil 2011 Singh et al., 2011 Singh et al., 2015 et al., Frey 2018 et al., Reschka 2010 et al., Bloemendal 2015 et al., Nordzieke 2014 Nordzieke,

Specificity domain anchor on tail Dependent (2 isoforms) on dependent localizations, Several B) kinase (protein Akt with interactions on and protein) DJ-1 (antioxidant and cellular stress other subunits all of Localization and on HAM3 (striatin) dependent HAM2 (STRIP) other subunits all of Localization Far10 and on Far9 dependent some of localization Asymmetric no interdependence components; partner MOB3 interaction and on PRO11 (striatin) Dependent PRO22 (STRIP) in Δ pro11

Localization Nuclear envelope Nuclear envelope ER, nuclear nuclear (during ER, centrosomes division) Nucleus/perinuclear and envelope/ER Nuclear mitochondria Autophagosomes ER probably system, Endomembrane (interphase), envelope Nuclear (mitosis) centrosomes Golgi PM ER, mitochondria, envelope Nuclear ER envelope Nuclear ring Actomyosin to (analogous body pole Spindle centrosome) mammalian Mitochondria envelope Nuclear tubular system, Intermembrane vacuoles mitochondria and envelope Nuclear bodies pole Spindle

Component STRIPAK Striatin (striatin) Farl11 STRIP2 SLMAP (striatin) Cka (striatin) Fsr1 SLMAP STRP1 (STRIP) STRN3 (striatin), SG2NA (striatin) STRIPAK SG2NA (striatin) homolog) (STRIPAK complex Far PP2A (Paa1) homolog) (STRIPAK complex SIP GPI1 (SIKE) SCI1 PRO11 (striatin), PRO22 (STRIP) PRO45 (SLMAP) PRO45 (SLMAP)

Localization of STRIPAK subunits. STRIPAK of 3: Localization Table Organism A. nidulans A. nidulans C. elegans (mouse) cells stem embryonic CGR8 kidney), monkey green (African COS7 (mouse) C2C12 cells melanogaster D. verticillioides F. (human) HeLa cells (human) HeLa cells (mouse) cells Neuro2A N. crassa (mouse) NIH3T3 cells cerevisiae S. pombe S. pombe S. macrospora S. macrospora S. macrospora S. macrospora S. macrospora S. U. Kück et al.: STRIPAK in development and disease 1011

STRIPAK controls MAPK localization acts as a negative regulator of tissue growth. Loss of func- tion in this kinase resulted in increased tissue growth in filamentous fungi and decreased apoptosis, and the corresponding mutants were named Hippopotamus because of their bulky build MAPK pathways transduce input signals to factors in the (Harvey et al., 2003; Wu et al., 2003). Later, the Hippo nucleus for altering gene transcription. For the filamen- kinase was identified as a part of a large signaling tous fungus Neurospora crassa, the STRIPAK complex is network, which is conserved from to eukaryotic known to localize to the nuclear envelope and regulate microbes (Pan, 2007, 2010). A fungal homolog of HIPPO is the nuclear import of MAPK MAK-1 in a MAK-2-dependent the septation initiation network (SIN), first discovered in manner (Dettmann et al., 2013). While MAK-2 (ERK homol- fission yeast S. pombe (Guertin et al., 2000; Simanis, 2015). ogous MAPK) is the homolog of the yeast pheromone Meanwhile, SIN homologs were described in various uni- response (PR) pathway MAPK, MAK-1 (JNK/p38 homolo- cellular and filamentous fungi. To compare designations gous MAPK) is a homolog of the yeast cell wall integrity of SIN subunits with those from fly and humans, Table 4 (CWI) pathway MAPK. In N. crassa, both kinases are provides a comprehensive overview of the literature and essential for cell-cell communication and fusion (Fischer designates synonymous subunits. et al., 2019). In humans, the Hippo pathway core consists of Similar results were recently obtained for A. nidu- GCK-II kinase MST1/2 (also known as STK4/3) associated lans. Homologs of MAK-1 and MAK-2 were investigated in with the WW-domain-containing scaffold protein Salva- mutants lacking distinct subunits of the STRIPAK complex dor SAV1 and the nuclear dbf2-related (NDR) kinase large (Elramli et al., 2019). From this set of data, an interest- tumor suppressor (LATS) 1/2, which is associated with ing mechanistic model was postulated that predicts that the kinase activator MOB1 (Pan, 2010) (Table 4). Recent STRIPAK promotes phosphorylation of MAK-2/ERK, a data provide compelling evidence of GCKs being one kinase of the PR pathway, and modulates its shuttling into of the major links between Hippo and STRIPAK. Five of the nucleus (Figure 3). There, the kinase phosphorylates eight animal GCK subfamilies (GCK-I, II, III, IV and VIII) transcription factors controlling processes such as sexual listed in Table 2 and graphically presented in Figure 4 and asexual development and the biosynthesis of second- were shown to functionally interact with either the Hippo ary metabolites. At the same time, nuclear transport of the pathway, the STRIPAK complex, or with both (Couzens CWI pathway MAPK MAK-1/JNK/p38 is constrained in the et al., 2013; Madsen et al., 2015; Meng et al., 2015; Thomp- absence of stress conditions, thereby maintaining proper son and Sahai, 2015; Meng et al., 2016). Hippo subunit stress signals in the cytoplasm. MOB1 was described as an interacting partner and a sub- strate of PP2A (Moreno et al., 2001) (Figure 4, pathway b). Structural and biochemical studies went on to discover a key mechanism of MST1/2 activation in which GCK-II Crosstalk between STRIPAK and kinases bind via their C-terminal linker to a forkhead- Hippo pathway associated (FHA) domain of the STRIPAK subunit SLMAP, thereby enabling their dephosphorylation by PP2Ac. SAV1 The Hippo pathway was first described in D. melanogaster. inhibits phosphatase activity of PP2A via binding to the Mutant screening in D. melanogaster identified a GCK that STRIPAK core, thereby counteracting dephosphorylation

Table 4: Components of the septation initiation network (SIN) complex, the homologue of the Hippo pathway in fly and humans.

Protein feature MEN SIN Hippo

Sc Sp An Nc Sm Dm Hs

STE kinase Cdc15 Cdc7 SEPH CDC-7 SMAC_04024 / / GCK / Sid1 SEPL SID1 SmKIN3, SMAC_04490 Hpo MST1 (STK4)/ MST2 (STK3) GCK adaptor / Cdc14 SEPM CDC-14 SMAC_06569 Sav? SAV1? NDR kinase Dbf2p Sid2 SIDB DBF-2 SMAC_05230 Wts Lats1/2 NDR kinase adaptor Mob1p Mob1 MOBA MOB-1 SMAC_02346 Mats MOB1

In baker’s yeast, the SIN complex was named mitotic exit network (MEN) (Lee et al., 2001) (Modified from Radchenko, 2018). An, A. nidulans; Dm, D. melanogaster; Hs, Homo sapiens; Nc, N. crassa; Sc, S. cerevisiae; Sm, S. macrospora; Sp, S. pombe. 1012 U. Kück et al.: STRIPAK in development and disease

STRIPAK P+ GCK-IV GCK-III MAP4K4/6 MST3/4, YSK1 STRIP CCM3 Striatin 1/2

P– MOB3 PP2AA PP2Ac i SLMAP I GCK-I MAP4K1/2/3/5 c P+ d

RASFF a GCK-VIII TAO1/2/3 SAV1 P– b k GCK-II P+ MST1/2 j f e P+ Hippo h P+ P+ MOB1 P– P+ MOB2 g NDR1/2 LATS1/2 P+ P+ P+

Figure 4: Signal modulation between STRIPAK complex and the GCK network. Positive (solid arrows), and negative regulations (blunt-end dashed lines), physical translocations (dotted lines), as well as phosphorylation and dephosphorylation reactions (P + and P-, respectively) are indicated. This model is based on Homo sapiens and D. melanogaster studies as referenced in this review. Different regulatory pathways are indicated by small letters (‘a’ to ‘k’). The STRIPAK complex emerges as a negative regulator of the Hippo pathway by dephosphorylating the kinase activation-loop of MST1/2 (A) and kinase activator MOB1 (B) (Moreno et al., 2001; Couzens et al., 2013). MST1/2 can interact with the STRIPAK complex via binding to an FHA domain of SLMAP (C). This interaction recruits SAV1 to the STRIPAK core and enables reciprocal negative regulation of its phosphatase activity (D), by a yet unknown mechanism (Bae et al., 2017). The STRIPAK/Hippo crosstalk is extended with GCK-III and GCK-IV subfamily kinases, which are able to phosphorylate NDR kinases NDR1/2 (E) and LATS1/2 (F), the targets of MST1/2 (G, H) (Stegert et al., 2005; Meng et al., 2015; Tang et al., 2015; Zheng et al., 2017; Meng et al., 2016). Additionally, GCK-III kinase MST3 might regulate the Hippo pathway by phosphorylating GCK- VIII kinase TAO1 (I), which in turn acts as MST1/2 activator (J) (Boggiano et al., 2011; Ultanir et al., 2014). Members of GCK-I subfamily were not identified as direct STRIPAK or Hippo core interaction partners, nevertheless they have shown the activity in LATS1/2 phosphorylation (K) (Meng et al., 2015; Zheng et al., 2017). Since the catalytic subunit of PP2A acts as a negative regulator of GCK-II (A) and GCK-III (L) subfamily kinases (Moreno et al., 2001; Gordon et al., 2011), it may be hypothesized that the STRIPAK complex also regulates other GCK group members via dephosphorylation. of the GCK-II activation loop. This mechanism is respon- Crosstalk between STRIPAK and sible for the reciprocal downregulation of STRIPAK and Hippo (Figure 4, pathways a, c, d) (Couzens et al., 2013; fungal homologs of the Hippo Bae et al., 2017). MST1/2 are capable of auto-activation pathway by self-phosphorylation but can also be activated by upstream regulatory GCK-VIII kinases TAO1/2/3, whose The core of SIN in fission yeast consists of the GCK Sid1p, phosphorylation state is dependent on the STRIPAK associated with a GCK adaptor Cdc14p, and an NDR kinase GCK-III kinases (Figure 4, pathways i, j) (Fallahi et al., Sid2p, associated with its activator Mob1p (Table 4). Dele- 2016; Meng et al., 2016). Hippo pathway dysfunction was tion or loss-of-function mutations of SIN members in S. described in mammalian cancer cell proliferation, migra- pombe result in a septation-deficient phenotype (Guertin tion and metastasis (Harvey et al., 2013). In particular, et al., 2000). Moreover, overexpression of SIN subunits, loss of function of the Hippo kinase homolog in human as well as deletion or loss of function of SIN negative cells is connected to carcinoma, adenoma and acute leu- regulators leads to hyperseptation (Simanis, 2015). The kemia (Pan, 2010; Harvey et al., 2013; Richardson and STRIPAK homolog in S. pombe is the SIN-inhibitory phos- Portela, 2017). phatase (SIP) complex, which acts as a negative regulator U. Kück et al.: STRIPAK in development and disease 1013 of SIN. Among other subunits, SIP comprises the striatin interact not only physically, but also genetically with the homolog Csc3p, PP2A scaffolding, and catalytic subunits striatin homolog PRO11 (Radchenko et al., 2018). Paa1p and Ppa3p, as well as the SLMAP homolog Csc1p (Table 1). Like its metazoan counterpart SLMAP, Csc1p acts as a link between SIP and SIN. However, in S. pombe, Ppa3p does not directly dephosphorylate a GCK activation Actin assembly is governed by the loop, but rather the SPB-targeted scaffold protein Cdc11p HIPPO/SIN and STRIPAK complexes (Tomlin et al., 2002). Cdc11p is required for the localiza- tion of other SIN components to SPBs and its dephospho- In eukaryotic cells, actin monomers exist as a free pool, rylation by SIP negatively regulates the SIN pathway. The each bound with one molecule of profilin and ATP. These dephosphorylation of SIN results in its inactivation and monomers can be polymerized in two different ways: dislocation from SPBs (Singh et al., 2011). In conclusion, either linear or branched. Generation of branched actin in S. pombe, SIN is negatively regulated by the STRIPAK filaments is governed by the actin-related protein (Arp) homolog SIP, which is comparable to downregulation of 2/3 complex, while linear polymerization is formin- Hippo through human STRIPAK. dependent (Insall and Machesky, 2009). Actin assembly In filamentous fungi, all subunits of the SIN three- is a major factor providing cell polarization, mechanistic step kinase cascade were recently identified in N. crassa support, cytokinesis and cell movement. and their hierarchy was confirmed by in vitro phospho- In humans, misregulation of actin assembly at any rylation assays (Table 4). GCK CDC-7, which is constitu- time causes defects in cell polarity and migration, which tively localized to SPBs, interacts with GCK SID-1 via the in turn are associated with different cancer types and neu- GCK adaptor CDC-14 and phosphorylates SID-1. Activated rodegenerative diseases (Insall and Machesky, 2009). In SID-1 then phosphorylates NDR kinase DBF-2, which is D. melanogaster, different types of actin polymerization associated with the kinase adaptor MOB-1 (Heilig et al., are required in different cell types for promoting a myriad 2013). Interestingly, CDC-7 can form a complex not only of key cellular and developmental processes. Finally, with SID-1, but also with GCK MST-1, and in a mutually during fungal development, it has been suggested that exclusive manner. Subsequently, the CDC-7-bound MST-1 different functions correspond to differently polymerized becomes enzymatically inactive. Active MST-1 is able to actin filaments. phosphorylate either of two NDR kinases, namely COT-1 In vertebrates and invertebrates, formins and the or DBF-2. These features highlight GCK MST-1 as a link Arp2/3 complex are filopodium formation regulators (Pel- between two NDR pathways – a phenomenon that is legrin and Mellor, 2005). Filopodia, also known as micro- similar to GCK-III kinases in metazoan (Figure 4E) (Heilig spikes, are thin cytoplasmic projections of migrating cells, et al., 2014). In N. crassa, the main function of SIN in the which appear when actively growing actin filaments push promotion of septation and cytokinesis was confirmed towards the plasma membrane. To date, two independ- by the phenotypes of SIN mutants. In detail, deletion ent mechanisms for filopodia formation are known. The of genes encoding CDC-7 and DBF-2 results in septation- first mechanism involves formin Dia2, activated by the deficient vegetative mycelium, while deletion mutants small GTPase Rho in filopodia (Rif), inducing the gen- lacking SID-1 and its adaptor CDC-14 generate aseptate eration of thin elongated microspikes on the cell surface. mycelium, which later reverts to a wildtype phenotype The second mechanism is driven by the Arp2/3 complex (Heilig et al., 2013). In A. nidulans, knowledge about SIN and Rho GTPase Cdc42. The Arp2/3 complex binds slow- has been mainly derived from the screening of temper- growing ends of daughter filaments and anchors them to ature-sensitive cytokinesis-deficient mutants (Harris the mother filament, thereby forming a highly branched et al., 1994). There, lack of the SIN NDR kinase SIDB structure that shapes the cell membrane into compact and its activator MOBA causes complete loss of septa- lamellipodia (Figure 5A). Overexpression of Rif in mam- tion and conidiation (Kim et al., 2006). In S. macrospora, malian cell culture results in the formation of elongated two STRIPAK-associated GCKs were described, namely cells with long and thin filopodia (Passey et al., 2004). SmKIN3 and SmKIN24 (Frey et al., 2015). Strains lacking In contrast, overexpression of Cdc42 results in rounded the Hippo-related GCK SmKIN3 exhibit rare septation and cells with short and thick filopodia. Remarkably, a similar early sexual development arrest. A more detailed analy- effect was observed in fibroblasts with depletion of either sis showed that strains carrying a point mutation in the Strip1 or Strip2. Of interest is that Strip1-deficient cells ATP-binding domain of SmKIN3 exhibit severe develop- tend to flat spreading and the shape of Strip2-deficient mental defects. More recently, SmKIN3 has been shown to cells is rather elongated, while both mutant cell lines 1014 U. Kück et al.: STRIPAK in development and disease

ABAnimals Fibroblast Neuronal axon Long thin filopodia Primary bouton (Dia2) (Rif) (Rif) STRIPAK (Dia2) STRIPAK Strip1 Hippo STRIPAK Cdc42 Strip2 Cdc42 Arp2/3 Arp2/3

Satellite Short thick boutons filopodia Filamentous fungi

C

Ascogonial septation

STRIPAK SISIN STRIPAK Trunk (RHO1) Linear (SepA) growth hyphae SIN septation

Branch base septation Cdc42 Arp2/3

Branching

Figure 5: SIN/Hippo and STRIPAK regulate actin cytoskeleton organization in animals and filamentous fungi. The model does not represent the localization of proteins/protein complexes, but only regulatory pathways. Arrows and blunt-end lines indicate activation and inhibition, respectively. Actin polymerization agents (Arp2/3 or Dia2/SepA) and associated (Cdc42 and Rif/RHO1) are indicated with blue and red font, respectively. Corresponding animal and fungal homologs with different nomenclature are enclosed in brackets. Actin structures, such as linear or branched filaments in animal cells, as well as actomyosin rings and actin cables in fungal cells are shown as dashed blue lines. show a decrease in migration. These findings suggest that fulfil the function of fine synaptic signal modulation. Of two STRIPAK sub-complexes, containing either Strip1 note is that in mutants with increased satellite bouton or Strip2, regulate actin cytoskeleton organization in an formation, abnormally decreased signaling levels are opposite manner, i.e. activating or inhibiting Rif-Dia2 observed. Overexpression of Arp2/3 or the Hippo kinase and Cdc42-Arp2/3 activities, respectively (Figure 5A; Bai as well as depletion of Strip causes formation of multiple et al., 2011). satellite boutons (Figure 5B). Moreover, knockdown of Another effect of Strip depletion is the malformation Hippo suppresses the Strip-deficient phenotype, thereby of the nervous system. In D. melanogaster, depletion of revealing the function of the STRIPAK complex as a nega- the single Strip homolog phenocopies overexpression of tive regulator of the Hippo pathway. Finally, Hippo kinase Arp2/3 during synapse development, which includes for- may consecutively phosphorylate and inactivate the actin mation of presynaptic termini (boutons) between motor polymerase Enabled (Ena), which antagonizes Arp2/3 and neuron and muscle cell. Besides primary boutons, which Cdc42 activity (Figure 5B; Sakuma et al., 2016). are always formed in wild-type neurons, generation of In fungi, proper actin assembly is essential for budding smaller satellite boutons is also possible. Satellite boutons and cytokinesis in yeast and polarized growth, branching emerge from the main nerve or bud from the primary and septation in filamentous fungi. During the process of bouton. During normal development, these satellites may septation, hyphal filaments are divided into compartments U. Kück et al.: STRIPAK in development and disease 1015 by cross-walls named septa. In most fungi, septa are perfo- arrest, the yeast Far (STRIPAK) complex is also known to rated by a large central pore that allows the movement of be involved in mitophagy. Mitophagy is a type of selective cytoplasm and organelles between cells. Septation occurs in autophagy that is responsible for mitochondrial quality and vegetative hyphae (trunk hyphae and lateral branches) as quantity control. Mitophagy defects cause various diseases, well as in sexual organs (ascogonia) (Figure 5C). Branching such as different cancer types and neurodegenerative dis- and septation in filamentous and dimorphic fungi are crucial eases. ATG32 is a mitophagy receptor in yeast that has to be for growth, nutrient acquisition, interaction with the envi- phosphorylated in a distinct manner to induce mitophagy. ronment, pathogenicity, as well as for asexual and sexual More recent studies have shown that the Far complex is propagation (Harris, 2008; Mouriño-Pérez, 2013; Riquelme responsible for the regulated dephosporylation of ATG32, et al., 2018). Unlike metazoan, fungi possess a solid cell thus counteracting the phosphorylation process and pre- wall, which maintains a rigid cell shape and does not allow venting extensive mitophagy (Furukawa et al., 2018). temporary protrusions or amoeboid motility. However, cell In the two related filamentous ascomycetes N. crassa wall protrusions in fungi can be observed during budding and S. macrospora, diverse (morphological) phenotypes or branching. The main branching regulator in A. nidulans were observed when mutants were investigated lacking and N. crassa is the conserved Rho GTPase Cdc42. In both distinct genes for STRIPAK subunits. The two most sig- ascomycetes, loss-of-function mutation of Cdc42 leads to nificant phenotypes observed were (1) lack of mature compromised growth, aberrant branching and abnormal fruiting bodies, rendering these mutants sterile, as well hyphal morphology (Virag et al., 2007; Araujo-Palomares as (2) their inability to generate hyphal (cellular) fusions et al., 2011). ARP-3 in N. crassa, the homolog of metazoan (Xiang et al., 2002; Pöggeler and Kück, 2004; Bloemendal Arp3, colocalizes with actin patches on future branch- et al., 2010; Simonin et al., 2010; Bernhards and Pögge- ing sites and follows the contour of the emerging branch ler, 2011; Fu et al., 2011; Dettmann et al., 2013). Most (Delgado-Alvarez­ et al., 2010). In this way, appearance of significantly, all STRIPAK mutants display identical phe- lateral branches resembles satellite bouton formation in notypes, indicating that all STRIPAK subunits function in neuronal cells from D. melanogaster (Figure 5B,C). Fungi concert (Teichert et al., 2014; Gautier et al., 2018; Elramli possess fewer formin paralogs than animals. Consequently, et al., 2019). Remarkably, in some filamentous fungi, such depletion of fungal formins leads to more severe defects. as Fusarium verticillioides and Epichloë festucae, even In S. pombe and S. cerevisiae, the homologs of the actin pathogenicity or symbiotic interactions are controlled by polymerizing agent Dia2 are Cdc12p and Bni1p, respectively. STRIPAK (Green et al., 2016; Zhang et al., 2018). These Dia2 homologs are required for the formation of the fungal-specific contractile actin ring as well as budding (Pollard, 2007). Filamentous ascomycetes, such as A. nidu- lans, N. crassa, and S. macrospora, only encode one formin, STRIPAK directs neuronal and SepA. Deletion of sepA is lethal in A. nidulans (Sharpless embryo development and Harris, 2002). Heterokaryotic strains of N. crassa with only some nuclei lacking the SepA homolog BNI-1 show As described, striatin is preferentially expressed in neurons severe hyphal growth defects as well as loss of septation in the striatum. However, little is as yet known about the (Lichius et al., 2012). Moreover, SepA was reported as a SIN precise function of striatin or the STRIPAK complex within phosphorylation target in A. nidulans (Sharpless and Harris, the neurobiological context. A recent shRNA knockdown 2002) and an interaction partner of the STRIPAK subunit approach in primary striatal neuronal cultures showed PRO45 in S. macrospora (Nordzieke et al., 2015). Collectively, the selective role for striatin in neuron maturation (Li these reports indicate a SIN/STRIPAK-dependent regulation et al., 2018). In detail, the knockdown approach resulted of septum formation in fungi (Figure 5C). in reduced expression of striatin, followed by an increase in dendritic complexity as well as density of dendritic spines. Thus, this study highlights the regulatory role of striatin in neuronal development. Further investigation of STRIPAK directs development in the interaction of STRIPAK with cortactin-binding protein eukaryotic microbes 2 (CBP2) has also demonstrated the role of STRIPAK subu- nits in neuronal development. CBP2 is a neuron-specific, In addition to the association of the STRIPAK complex F-actin-associated protein that regulates the formation with conserved signaling pathways, such as controlling and maintenance of dendritic spines. CBP2 has previously yeast cellular processes and cell wall integrity or cell cycle been associated with autistic spectrum disorder and was 1016 U. Kück et al.: STRIPAK in development and disease suggested to be critical for neural development and func- known about the role of STRIPAK complexes during mam- tion (Chen et al., 2012). Using imaging analysis of cultured malian development in general and pertaining underlying hippocampal neurons, these authors also predicted that molecular mechanisms in particular. Notably, however, CBP2 targets STRIPAK to dendritic spines. recent investigations have indicated that the dysregula- Another example of the developmental role of a tion of subunits of the STRIPAK complexes correlates with STRIPAK subunit was recently demonstrated for Strip1. In numerous human diseases, including different cancer mouse embryos, a Strip1 mutation disrupts migration of the types and diabetes as well as cardiovascular and fatty mesoderm after the gastrulation ­epithelial-to-mesenchymal liver diseases. A compilation is provided in Table 5. transition (EMT). Complete deletion of the gene for Strip1 In mammalian cells, the scaffold subunit of the resulted in profound disruptions in the organization of STRIPAK protein phosphatase 2A (PP2AA) exists in two the mesoderm and its derivatives. Finally, these studies isoforms (α/β), for which mutations in the corresponding also revealed that the mesoderm migration defect is corre- genes correlate with different forms of human carcinomas lated with changes in actin cytoskeleton organization and (Calin et al., 2000). These mutations include point muta- decreased velocity of cell migration (Bazzi et al., 2017). tions as well as exon deletions and occurred at low fre- In Drosophila, the CLOCK/CYCLE transcription quency. Mutations in other STRIPAK subunits also seem to factors activate the transcription of period and timeless correlate with distinct disease phenotypes. For example, genes, which are essential components of the molecular in boxer , a deletion in the striatin gene was associ- clock controlling circadian rhythms. The CLOCK protein ated with dilated cardiomyopathy (DCM) and arrhythmo- is highly phosphorylated and inactive in the morning, genic right ventricular cardiomyopathy (ARVC) (Meurs whereas hypophosphorylated active forms are present in et al., 2013). Similarly, polymorphic variants of the human the evening. The phosphorylation status is highly depend- striatin (STRN1) gene have been associated with increased ent on subunits of the Drosophila STRIPAK complex, salt sensitivity of blood pressure. These results were namely the regulatory striatin subunit of PP2A and the further confirmed by knockdown experiments using the interacting STRIP subunit (Andreazza et al., 2015). mouse striatin gene (Garza et al., 2015). Likewise, silenc- ing of the striatin (STRN4) gene in mouse cell cultures sup- presses proliferation, migration, and invasion of cancer cells (Wong et al., 2014). More recently, another transcrip- Human and mammalian diseases tionally controlled mechanism was predicted when RNA from patients showing intracranial aneurysm was used in In recent years, the role of STRIPAK complexes in cellular association studies. Of note is that among downregulated and developmental processes of multiple model organisms genes in these samples was striatin (STRN). The authors has been extensively studied. However, little is currently therefore suggested that striatin may be associated with

Table 5: Mammalian diseases correlated with dysfunction of STRIPAK subunits.

STRIPAK subunit Gene change Disease/syndrome Organism Reference

PP2AAα/β E64D Lung carcinoma Human Calin et al., 2000 E64G; frame shift Breast carcinoma R418W Melanoma Alternative splicing, exon Breast carcinoma 9 skipping Striatin1 Downregulated Intracranial aneurysm (IA) Human Wei et al., 2018 8 bp deletion in the 3′ UTR Dilated cardiomyopathy (DCM), arrhythmogenic Meurs et al., 2013 right ventricular cardiomyopathy (ARVC) G/A point mutation 5′ UTR Salt-sensitive hypertension Human Garza et al., 2015 SG2NA/Striatin3 Upregulated Cancer and metastasis development Mouse Wong et al., 2014 YSK1 Upregulated Type 2 diabetes, non-alcoholic fatty liver Mouse Amrutkar et al., 2016; disease Chursa et al., 2017 MST4 Upregulated Prostate cancer and pituitary tumorigenesis Human Sung et al., 2003; Xiong et al., 2015 SLMAP V269I, E710A and some Brugada syndrome/cardiac channelopathy Human (Asians) Ishikawa et al., 2012 other SNPs U. Kück et al.: STRIPAK in development and disease 1017 the development of intracranial aneurysm via influencing controls many cellular and organismal processes, includ- the development of neuron projections of the correspond- ing immune and inflammatory responses, cellular growth, ing cells (Wei et al., 2018). and cell survival. Studies with protein phosphatase holo- RNA association studies were also conducted to PP2A showed that only the regulatory subunit B′′′/ study the function of STRIPAK-associated kinases. Over- striatin promotes the dephosphorylation of all three subu- expression and knockdown experiments with the YSK1 nits of NF-κB (Tsuchiya et al., 2017). In this context, it is (synonymous to STK25) gene highlighted its role in type interesting that transactivation of the glucocorticoid recep- 2 diabetes and non-alcoholic fatty liver diseases (Amrut- tor (GR) , which regulates genes con- kar et al., 2016; Chursa et al., 2017). In follow-up studies trolling development, metabolism, and immune response, using YSK1 transgenic and wild-type mice, a global quan- depends on striatin (STRN3). STRN3 itself is a prerequi- titative phosphoproteomic analysis identified 21 proteins site for the formation of a complex between the catalytic that were differentially phosphorylated, indicating these subunit of protein phosphatase 2A (PP2AC) with GR, which as potential downstream mediators of STRIPAK action results in its downregulation. It was hypothesized that a (Chursa et al., 2017). functional trimeric protein phosphatase 2A complex in Similar association studies were also performed with the nucleus dephosphorylates GR at serine 211, a known the STRIPAK-related MST4 (synonymous to STK23) kinase. marker for GR transactivation (Petta et al., 2017). Ectopic expression of the wild-type and kinase-inactive Another upstream regulatory mechanism was recently MST4 gene in human prostate tumor cell lines showed described when 17β-estradiol (E2) was investigated, which that overexpression of MST4 increases proliferation and has a beneficial effect on the cardiovascular system. E2, tumorigenesis, while downregulation of this gene reverts which induces the phosphorylation of phosphoinositide this highly tumorigenic behavior. Taken together, these 3-kinases (PI3Ks), increases striatin protein expression in studies suggest a potential role of MST4 in signaling path- a dose- and time-dependent manner in human umbilical ways involved in prostate cancer progression. Moreover, vein endothelial cells (HUVECs). Interestingly, the treat- microarray expression analysis with samples from human ment of HUVECs with the phosphatidylinositol-3 kinase tumors revealed that the MST4 gene is upregulated in all inhibitor, wortmannin, abolished E2-mediated upregula- gonadotrope tumor samples investigated. Thus, MST4 tion of striatin protein expression. Wortmannin, a fungal seems to be a novel candidate that is involved in human steroid metabolite, is a covalent inhibitor of PI3Ks, which pituitary tumorigenesis (Xiong et al., 2015). are recruited to the membrane and that act upstream of The STRIPAK subunit SLMAP is also known to be the STRIPAK pathway. Thus, E2 seems to regulate striatin involved in pathophysiological processes. For example, via the PI3K signal transduction pathway (Zheng et al., association studies with samples from Asian patients 2018). Collectively, the abovementioned examples provide showed that SLMAP mutations have an effect on cell the first mechanistic insights on how STRIPAK dysfunc- surface expression of the pore-forming α-subunit of the tion might cause human diseases on the cellular level. cardiac sodium channel hNav1.5 (Ishikawa et al., 2012). The corresponding gene is also known to underlie Brugada syndrome, a cardiac channelopathy, which is often accom- panied by syncope and sudden cardiac death attributable Future perspectives to ventricular arrhythmias (Brugada and Brugada, 1992). The conclusion that SLMAP plays a pivotal role in Brugada Since the first description of striatin about 20 years ago, syndrome was further substantiated by small interfering striatin and its associated proteins have been further RNA experiments and whole-cell patch clamp recordings investigated in a broad range of studies using diverse (Ishikawa et al., 2012). eukaryotic experimental and model systems. Subse- Thus, despite the accumulating evidence that STRIPAK quently, several structural and mechanistic models have subunits are linked with mammalian diseases, we currently been proposed to explain the role of the STRIPAK complex have very little mechanistic understanding of STRIPAK in diverse cellular and developmental processes occurring function in pathogenicity. New insights into mechanistic in both microbial as well as animal systems. Despite enor- action may come from studies with cell cultures investi- mous progress in biochemically characterizing STRIPAK gating different forms of PP2A phosphatase that regulates complexes, key questions still remain about the regula- the NF-κB pathway. This pathway consists of transcription tion of developmental processes as well as the cross-talk factor nuclear factor kappa B (NF-κB), together with at between STRIPAK and other conserved signaling path- least three of its subunits (IKKβ, IκBα, and RelA). NF-κB ways. For example, the mechanisms by which upstream 1018 U. Kück et al.: STRIPAK in development and disease regulators and downstream targets affect signaling of the Bernhards, Y. and Pöggeler, S. (2011). The phocein homologue STRIPAK complex are not yet fully understood. In particu- SmMOB3 is essential for vegetative cell fusion and sexual development in the filamentous ascomycete Sordaria macros- lar, it is unknown how the inactivation or inhibition of pora. Curr. Genet. 57, 133–149. subunits of the STRIPAK complex influence the phospho- Bloemendal, S., Lord, K.M., Rech, C., Hoff, B., Engh, I., Read, N.D., rylation status of target proteins. and Kück, U. (2010). A mutant defective in sexual development Phosphoproteomic studies may advance our current produces aseptate ascogonia. Eukaryotic Cell 9, 1856–1866. rudimentary understanding of how STRIPAK leads to the Bloemendal, S., Bernhards, Y., Bartho, K., Dettmann, A., Voigt, O., deregulation of cellular division processes in microbes or Teichert, I., Seiler, S., Wolters, D.A., Pöggeler, S., and Kück, U. (2012). A homologue of the human STRIPAK complex controls to the onset of diverse cancer types in mammals. Thus, sexual development in fungi. Mol. Microbiol. 84, 310–323. gaining additional insights into its upstream regulators Boggiano, J.C., Vanderzalm, P.J., and Fehon, R.G. (2011). Tao-1 phos- and downstream targets is crucial as a prelude to translat- phorylates Hippo/MST kinases to regulate the Hippo-Salvador- ing our basic knowledge of this complex and its signaling Warts tumor suppressor pathway. Dev. Cell 21, 888–895. pathways into therapeutic interventions. Given the rel- Boyce, K.J. and Andrianopoulos, A. (2011). Ste20-related kinases: effectors of signaling and morphogenesis in fungi. Trends evance of the STRIPAK complex for human diseases, such Microbiol. 19, 400–410. as cancer, this signaling pathway may thus provide prom- Brugada, P. and Brugada, J. (1992). Right bundle branch block, ising drug targets for pharmaceutical development aimed persistent ST segment elevation and sudden cardiac death: a at producing new therapies for distinct types of cancer. distinct clinical and electrocardiographic syndrome: a multi- center report. J. Am. Coll. Cardiol 20, 1391–1396. Acknowledgments: The experimental work of the authors Byers, J.T., Guzzo, R.M., Salih, M., and Tuana, B.S. (2009). Hydro- phobic profiles of the tail anchors in SLMAP dictate subcellular is funded by the German Science Foundation DFG targeting. BMC Cell Biol. 10, 48. (KU517/11-2, KU517/16-1, TE977/2-1), and Daria Radchenko Calin, G.A., Di Iasio, M.G., Caprini, E., Vorechovsky, I., Natali, P.G., received a scholarship from the Friedrich Ebert-Stiftung Sozzi, G., Croce, C.M., Barbanti-Brodano, G., and Russo, G. (Germany). We thank G. Frenßen-Schenkel for graphical (2000). Low frequency of alterations of the α (PPP2R1A) and assistance. β (PPP2R1B) isoforms of the subunit A of the serine-threonine phosphatase 2A in human neoplasms. Oncogene 19, 1191–1195. Castets, F., Bartoli, M., Barnier, J.V., Baillat, G., Salin, P., Moqrich, A., Bourgeois, J.P., Denizot, F., Rougon, G., Calothy, G., et al. References (1996). A novel calmodulin-binding protein, belonging to the WD-repeat family, is localized in dendrites of a subset of CNS Amrutkar, M., Kern, M., Nuñez-Durán, E., Ståhlman, M., Cansby, E., neurons. J. Cell Biol. 134, 1051–1062. Chursa, U., Stenfeldt, E., Borén, J., Blüher, M., and Mahlapuu, Chen, H.W., Marinissen, M.J., Oh, S.W., Chen, X., Melnick, M., M. (2016). Protein kinase STK25 controls lipid partitioning in ­Perrimon, N., Gutkind, J.S., and Hou, S.X. (2002). CKA, a novel hepatocytes and correlates with liver fat content in humans. multidomain protein, regulates the Jun N-terminal kinase Diabetologia 59, 341–353. signal transduction pathway in Drosophila. Mol. Cell Biol. 22, Andreazza, S., Bouleau, S., Martin, B., Lamouroux, A., Ponien, P., 1792–1803. Papin, C., Chélot, E., Jacquet, E., and Rouyer, F. (2015). Daytime Chen, Y.K., Chen, C.Y., Hu, H.T., and Hsueh, Y.P. (2012). CTTNBP2, but CLOCK dephosphorylation is controlled by STRIPAK complexes not CTTNBP2NL, regulates dendritic spinogenesis and synaptic in Drosophila. Cell Rep. 11, 1266–1279. distribution of the striatin-PP2A complex. Mol. Biol. Cell 23, Araujo-Palomares, C.L., Richthammer, C., Seiler, S., and Castro- 4383–4392. Longoria, E. (2011). Functional characterization and cellular Chen, C., Shi, Z., Zhang, W., Chen, M., He, F., Zhang, Z., Wang, Y., dynamics of the CDC-42-RAC-CDC-24 module in Neurospora Feng, M., Wang, W., Zhao, Y., et al. (2014). Striatins contain a crassa. PLoS One 6, e27148. noncanonical coiled coil that binds protein phosphatase 2AA Bae, S.J., Ni, L., Osinski, A., Tomchick, D.R., Brautigam, C.A., and subunit to form a 2:2 heterotetrameric core of striatin-interact- Luo, X. (2017). SAV1 promotes Hippo kinase activation through ing phosphatase and kinase (STRIPAK) complex. J. Biol. Chem. antagonizing the PP2A phosphatase STRIPAK. eLife 6, e30278. 289, 9651–9661. Bai, S.W., Herrera-Abreu, M.T., Rohn, J.L., Racine, V., Tajadura, V., Chursa, U., Nuñez-Durán, E., Cansby, E., Amrutkar, M., Sütt, S., Suryavanshi, N., Bechtel, S., Wiemann, S., Baum, B., and Ståhlman, M., Olsson, B.M., Borén, J., Johansson, M.E., Ridley, A.J. (2011). Identification and characterization of a set of ­Bäckhed, F., et al. (2017). Overexpression of protein kinase conserved and new regulators of cytoskeletal organization, cell STK25 in mice exacerbates ectopic lipid accumulation, morphology and migration. BMC Biol. 9, 54. mitochondrial dysfunction and insulin resistance in skeletal Bartlett, K. and Kim, K. (2014). Insight into Tor2, a budding yeast muscle. Diabetologia 60, 553–567. microdomain protein. Eur. J. Cell Biol. 93, 87–97. Couzens, A.L., Knight, J.D., Kean, M.J., Teo, G., Weiss, A., Dunham, Bazzi, H., Soroka, E., Alcorn, H.L., and Anderson, K.V. (2017). STRIP1, W.H., Lin, Z.Y., Bagshaw, R.D., Sicheri, F., Pawson, T., et al. a core component of STRIPAK complexes, is essential for (2013). Protein interaction network of the mammalian Hippo normal mesoderm migration in the mouse embryo. Proc. Natl. pathway reveals mechanisms of kinase-phosphatase interac- Acad. Sci. USA 114, E10928–E10936. tions. Sci. Signal 6, rs15. U. Kück et al.: STRIPAK in development and disease 1019

Delgado-Alvarez, D.L., Callejas-Negrete, O.A., Gómez, N., Freitag, sity interaction network identifies a novel striatin-interacting M., Roberson, R.W., Smith, L.G., and Mouriño-Pérez, R.R. phosphatase and kinase complex linked to the cerebral cavern- (2010). Visualization of F-actin localization and dynamics with ous malformation 3 (CCM3) protein. Mol. Cell. Proteomics 8, live cell markers in Neurospora crassa. Fungal. Genet. Biol. 47, 157–171. 573–586. Green, K.A., Becker, Y., Fitzsimons, H.L., and Scott, B. (2016). An Delpire, E. (2009). The mammalian family of sterile 20p-like protein Epichloë festucae homologue of MOB3, a component of the kinases. Pflüger’s Arch. 458, 953–967. STRIPAK complex, is required for the establishment of a mutu- Dettmann, A., Heilig, Y., Ludwig, S., Schmitt, K., Illgen, J., Fleissner, alistic symbiotic interaction with Lolium perenne. Mol. Plant A., Valerius, O., and Seiler, S. (2013). HAM-2 and HAM-3 are Pathol. 17, 1480–1492. central for the assembly of the Neurospora STRIPAK complex Guertin, D.A., Chang, L., Irshad, F., Gould, K.L., and McCollum, D. at the nuclear envelope and regulate nuclear accumulation of (2000). The role of the sid1p kinase and cdc14p in regulating the MAP kinase MAK-1 in a MAK-2-dependent manner. Mol. the onset of cytokinesis in fission yeast. EMBO J. 19, 1803–1815. Microbiol. 90, 796–812. Harris, S.D. (2008). Branching of fungal hyphae: regulation, Elramli, N., Karahoda, B., Sarikaya Bayram, Ö., Frawley, D., Ulas, M., mechanisms and comparison with other branching systems. Oakley, C.E., Oakley, B.R., Seiler, S., and Bayram, Ö. (2019). Mycologia 100, 823–832. Assembly of a heptameric STRIPAK complex is required for Harris, S.D., Morrell, J.L., and Hamer, J.E. (1994). Identification and coordination of light dependent multicellular fungal develop- characterization of Aspergillus nidulans mutants defective in ment with secondary metabolism in Aspergillus nidulans. PLoS cytokinesis. Genetics 136, 517–532. Genet 15, e1008053. Harvey, K.F., Pfleger, C.M., and Hariharan, I.K. (2003). The Drosoph- Fallahi, E., O’Driscoll, N.A., and Matallanas, D. (2016). The MST/ ila MST ortholog, Hippo, restricts growth and cell proliferation Hippo pathway and cell death: a non-canonical affair. Genes and promotes apoptosis. Cell 114, 457–467. (Basel) 7, pii: E28. Harvey, K.F., Zhang, X., and Thomas, D.M. (2013). The Hippo path- Fischer, M.S., Jonkers, W., and Glass, N.L. (2019). Integration of self way and human cancer. Nat. Rev. Cancer 13, 246–257. and non-self recognition modulates asexual cell-to-cell com- Heilig, Y., Schmitt, K., and Seiler, S. (2013). Phospho-regulation of munication in Neurospora crassa. Genetics 211, 1255–1267. the Neurospora crassa septation initiation network. PLoS One Frey, S., Lahmann, Y., Hartmann, T., Seiler, S., and Pöggeler, S. 8, e79464. (2015). Deletion of Smgpi1 encoding a GPI-anchored protein Heilig, Y., Dettmann, A., Mouriño-Pérez, R.R., Schmitt, K., Valerius, suppresses sterility of the STRIPAK mutant ΔSmmob3 in the O., and Seiler, S. (2014). Proper actin ring formation and filamentous ascomycete Sordaria macrospora. Mol. Microbiol. septum constriction requires coordinated regulation of SIN and 97, 676–697. MOR pathways through the germinal centre kinase MST-1. PLoS Frost, A., Elgort, M.G., Brandman, O., Ives, C., Collins, S.R., Genet. 10, e1004306. ­Miller-Vedam, L., Weibezahn, J., Hein, M.Y., Poser, I., Mann, Hwang, J. and Pallas, D.C. (2014). STRIPAK complexes: structure, M., et al. (2012). Functional repurposing revealed by compar- biological function, and involvement in human diseases. Int. J. ing S. pombe and S. cerevisiae genetic interactions. Cell 149, Biochem. Cell Biol. 47, 118–148. 1339–1352. Insall, R.H. and Machesky, L.M. (2009). Actin dynamics at the lead- Fu, C., Iyer, P., Herkal, A., Abdullah, J., Stout, A., and Free, S.J. ing edge: from simple machinery to complex networks. Dev. (2011). Identification and characterization of genes required Cell 17, 310–322. for cell-to-cell fusion in Neurospora crassa. Eukaryot. Cell 10, Ishikawa, T., Sato, A., Marcou, C.A., Tester, D.J., Ackerman, M.J., 1100–1109. Crotti, L., Schwartz, P.J., On, Y.K., Park, J.E., Nakamura, K., et al. Furukawa, K., Fukuda, T., Yamashita, S.I., Saigusa, T., Kurihara, Y., (2012). A novel disease gene for Brugada syndrome: sarcolem- Yoshida, Y., Kirisako, H., Nakatogawa, H., and Kanki, T. (2018). mal membrane-associated protein gene mutations impair intra- The PP2A-like protein phosphatase Ppg1 and the Far complex cellular trafficking of hNav1. 5. Circ. Arrhythm. Electrophysiol. cooperatively counteract CK2-mediated phosphorylation of 5, 1098–1107. Atg32 to inhibit mitophagy. Cell Rep. 23, 3579–3590. Jain, B.P., Pandey, S., Saleem, N., Tanti, G.K., Mishra, S., and Garza, A.E., Rariy, C.M., Sun, B., Williams, J., Lasky-Su, J., Baudrand, Goswami, S.K. (2017). SG2NA is a regulator of endoplasmic R., Yao, T., Moize, B., Hafiz, W.M., Romero, J.R., et al. (2015). reticulum (ER) homeostasis as its depletion leads to ER stress. Variants in striatin gene are associated with salt-sensitive Cell Stress Chaperones 22, 853–866. blood pressure in mice and humans. Hypertension 65, 211–217. Kemp, H.A. and Sprague, G.F. Jr. (2003). Far3 and five interacting Gautier, V., Tong, L.C.H., Nguyen, T.S., Debuchy, R., and Silar, P. proteins prevent premature recovery from pheromone arrest in (2018). PaPro1 and IDC4, Two genes controlling stationary the budding yeast Saccharomyces cerevisiae. Mol. Cell. Biol. phase, sexual development and cell degeneration in Podos- 23, 1750–1763. pora anserina. J. Fungi (Basel). 4, pii: E85. Kim, J.M., Lu, L., Shao, R., Chin, J., and Liu, B. (2006). Isolation of Gordon, J., Hwang, J., Carrier, K.J., Jones, C.A., Kern, Q.L., Moreno, mutations that bypass the requirement of the septation initia- C.S., Karas, R.H., and Pallas, D.C. (2011). Protein phosphatase tion network for septum formation and conidiation in Aspergil- 2a (PP2A) binds within the oligomerization domain of striatin lus nidulans. Genetics 173, 685–696. and regulates the phosphorylation and activation of the mam- Kück, U., Beier, A., and Teichert, I. (2016). The composition and malian Ste20-Like kinase Mst3. BMC Biochem. 12, 54. function of the striatin-interacting phosphatases and kinases Goudreault, M., D’Ambrosio, L.M., Kean, M.J., Mullin, M.J., Larsen, (STRIPAK) complex in fungi. Fungal Genet Biol 90, 31–38. B.G., Sanchez, A., Chaudhry, S., Chen, G.I., Sicheri, F., Kyriakis, J.M. (1999). Signaling by the germinal center kinase family ­Nesvizhskii, A.I., et al. (2009). A PP2A phosphatase high den- of protein kinases. J. Biol. Chem. 274, 5259–5262. 1020 U. Kück et al.: STRIPAK in development and disease

Lee, S.E., Frenz, L.M., Wells, N.J., Johnson, A.L., and Johnston, L.H. tal role in development and localizes to the nuclear envelope, (2001). Order of function of the budding-yeast mitotic exit- ER, and mitochondria. Eukaryot. Cell 14, 345–358. network proteins Tem1, Cdc15, Mob1, Dbf2, and Cdc5. Curr. Pan, D. (2007). Hippo signaling in organ size control. Genes Dev. 21, Biol. 11, 784–788. 886–897. Li, D., Musante, V., Zhou, W.L., Picciotto, M.R., and Nairn, A.C. Pan, D. (2010). The hippo signaling pathway in development and (2018). Striatin-1 is a B subunit of protein phosphatase PP2A cancer. Dev. Cell 19, 491–505. that regulates dendritic arborization and spine development in Passey, S., Pellegrin, S., and Mellor, H. (2004). What is in a filopo- striatal neurons. J. Biol. Chem. 293, 11179–11194. dium? Starfish versus hedgehogs. Biochem. Soc. Trans. 32, Lichius, A., Yanez-Gutierrez, M.E., Read, N.D., and 1115–1117. ­Castro-Longoria, E. (2012). Comparative live-cell imaging Pellegrin, S. and Mellor, H. (2005). The Rho family GTPase Rif analyses of SPA-2, BUD-6 and BNI-1 in Neurospora crassa induces filopodia through mDia2. Curr. Biol. 15, 129–133. reveal novel features of the filamentous fungal polarisome. Petta, I., Bougarne, N., Vandewalle, J., Dejager, L., Vandevyver, S., PLoS One 7, e30372. Ballegeer, M., Desmet, S., Thommis, J., De Cauwer, L., Lievens, Madsen, C.D., Hooper, S., Tozluoglu, M., Bruckbauer, A., Fletcher, S., et al. (2017). Glucocorticoid receptor-mediated transactiva- G., Erler, J.T., Bates, P.A., Thompson, B., and Sahai, E. (2015). tion is hampered by striatin-3, a novel interaction partner of STRIPAK components determine mode of cancer cell migration the receptor. Sci. Rep. 7, 8941. and metastasis. Nat. Cell. Biol. 17, 68–80. Pöggeler, S. and Kück, U. (2004). A WD40 repeat protein regulates Maheshwari, R., Pushpa, K., and Subramaniam, K. (2016). A role for fungal cell differentiation and can be replaced functionally post-transcriptional control of endoplasmic reticulum dynamics by the mammalian homologue striatin. Eukaryot. Cell 3, and function in C. elegans germline stem cell maintenance. 232–240. Development 143, 3097–3108. Pollard, T.D. (2007). Regulation of actin filament assembly by Arp2/3 Meng, Z., Moroishi, T., Mottier-Pavie, V., Plouffe, S.W., Hansen, complex and formins. Annu. Rev. Biophys. Biomol. Struct. 36, C.G., Hong, A.W., Park, H.W., Mo, J.S., Lu, W., Lu, S., et al. 451–477. (2015). MAP4K family kinases act in parallel to MST1/2 to Pracheil, T. and Liu, Z. (2013). Tiered assembly of the yeast Far3-7- activate LATS1/2 in the Hippo pathway. Nat. Commun. 6, 8-9-10-11 complex at the endoplasmic reticulum. J. Biol. Chem. 8357. 288, 16986–16997. Meng, Z., Moroishi, T., and Guan, K.-L. (2016). Mechanisms of Hippo Pracheil, T., Thornton, J., and Liu, Z. (2012). TORC2 signaling is pathway regulation. Genes Dev. 30, 1–17. antagonized by protein phosphatase 2A and the Far complex in Meurs, K.M., Stern, J.A., Sisson, D.D., Kittleson, M.D., Cunningham, Saccharomyces cerevisiae. Genetics 190, 1325–1339. S.M., Ames, M.K., Atkins, C.E., DeFrancesco, T., Hodge, T.E., Radchenko, D. (2018). Cellular signaling pathways in the filamentous Keene, B.W., et al. (2013). Association of dilated cardiomyopa- fungus Sordaria macrospora: Molecular Genetic Analysis of the thy with the striatin mutation genotype in boxer dogs. J. Vet. STRIPAK-Associated Germinal Center Kinase SmKIN3 in Develop- Intern. Med. 27, 1437–1440. mental Processes. PhD thesis (Ruhr-University Bochum). Moreno, C.S., Park, S., Nelson, K., Ashby, D., Hubalek, F., Lane, Radchenko, D., Teichert, I., Pöggeler, S., and Kück, U. (2018). A W.S., and Pallas, D.C. (2000). WD40 repeat proteins striatin Hippo pathway-related GCK controls both sexual and vegetative and S/G(2) nuclear autoantigen are members of a novel family developmental processes in the fungus Sordaria macrospora. of calmodulin-binding proteins that associate with protein Genetics 210, 137–153. phosphatase 2A. J. Biol. Chem. 275, 5257–5263. Reschka, E., Nordzieke, S., Valerius, O., Braus, G.H., and Pögge- Moreno, C.S., Lane, W.S., and Pallas, D.C. (2001). A mammalian ler, S. (2018). A novel STRIPAK complex component mediates homolog of yeast MOB1 is both a member and a putative sub- hyphal fusion and fruiting-body development in filamentous strate of striatin family-protein phosphatase 2A complexes. J. fungi. Mol. Microbiol. 110, 513–532. Biol. Chem. 276, 24253–24260. Ribeiro, P.S., Josué, F., Wepf, A., Wehr, M.C., Rinner, O., Kelly, Mouriño-Pérez, R.R. (2013). Septum development in filamentous G., Tapon, N., and Gstaiger, M. (2010). Combined functional ascomycetes. Fungal Biol. Rev. 27, 1–9. genomic and proteomic approaches identify a PP2A complex Neisch, A.L., Neufeld, T.P., and Hays, T.S. (2017). A STRIPAK as a negative regulator of Hippo signaling. Mol. Cell 39, complex mediates axonal transport of autophagosomes and 521–534. dense core vesicles through PP2A regulation. J. Cell Biol. 216, Richardson, H.E. and Portela, M. (2017). Tissue growth and tumo- 441–461. rigenesis in Drosophila: cell polarity and the Hippo pathway. Nguyen, T.A., Cissé, O.H., Yun Wong, J., Zheng, P., Hewitt, D., Curr. Opin. Cell Biol. 48, 1–9. Nowrousian, M., Stajich, J.E., and Jedd, G. (2018). Innovation Riquelme, M., Aguirre, J., Bartnicki-García, S., Braus, G.H., Feld- and constraint leading to complex multicellularity in the Asco- brügge, M., Fleig, U., Hansberg, W., Herrera-Estrella, A., Kämper, mycota. Nat. Commun. 8, 14444. J., Kück, U., et al. (2018). Fungal morphogenesis, from the polar- Nordzieke, S. (2014). Molecular genetic studies of PRO45 in Sord- ized growth of hyphae to complex reproduction and infection aria macrospora: a homolog of the sarcolemmal membrane- structures. Microbiol. Mol. Biol. Rev. 82, pii: e00068-17. associated protein (SLMAP) localizes to the nuclear envelope Sabour, D., Srinivasan, S.P., Rohani, S., Wagh, V., Gaspar, J.A., and plays an essential role in fungal development. PhD thesis Panek, D., Ardestani, M.A., Doss, M.X., Riet, N., Abken, H., (Ruhr-University Bochum). et al. (2017). STRIP2 is indispensable for the onset of embry- Nordzieke, S., Zobel, T., Fränzel, B., Wolters, D.A., Kück, U., and onic stem cell differentiation. Mol. Ther. Methods Clin. Dev. 5, Teichert, I. (2015). A fungal SLMAP homolog plays a fundamen- 116–129. U. Kück et al.: STRIPAK in development and disease 1021

Sakuma, C., Saito, Y., Umehara, T., Kamimura, K., Maeda, N., Mosca, Tomlin, G.C., Morrell, J.L., and Gould, K.L. (2002). The spindle pole T.J., Miura, M., and Chihara, T. (2016). The Strip-Hippo pathway body protein Cdc11p links Sid4p to the fission yeast septation regulates synaptic terminal formation by modulating actin initiation network. Mol. Biol. Cell. 13, 1203–1214. organization at the Drosophila neuromuscular synapses. Cell Tsuchiya, Y., Osaki, K., Kanamoto, M., Nakao, Y., Takahashi, E., Rep. 16, 2289–2297. Higuchi, T., and Kamata, H. (2017). Distinct B subunits of PP2A Sents, W., Ivanova, E., Lambrecht, C., Haesen, D., and Janssens, V. regulate the NF-κB signalling pathway through dephosphoryla- (2012). The biogenesis of active protein phosphatase 2A holo- tion of IKKβ, IκBα and RelA. FEBS Lett. 591, 4083–4094. : a tightly regulated process creating phosphatase Ultanir, S.K., Yadav, S., Hertz, N.T., Oses-Prieto, J.A., Claxton, S., specificity. FEBS J. 280, 644–661. Burlingame, A.L., Shokat, K.M., Jan, L.Y., and Jan, Y.N. (2014). Sharpless, K.E. and Harris, S.D. (2002). Functional ­characterization MST3 kinase phosphorylates TAO1/2 to enable Va and localization of the Aspergillus nidulans formin SEPA. function in promoting spine synapse development. Neuron 84, Mol. Biol. Cell. 13, 469–479. 968–982. Shi, Z., Jiao, S., and Zhou, Z. (2016). STRIPAK complexes in cell Virag, A., Lee, M.P., Si, H., and Harris, S.D. (2007). Regulation of signaling and cancer. Oncogene 35, 4549–4557. hyphal morphogenesis by and homologues in Simanis, V. (2015). Pombe’s thirteen – control of fission yeast Aspergillus nidulans. Mol. Microbiol. 66, 1579–1596. cell division by the septation initiation network. J. Cell Sci. Wang, C.L., Shim, W.B., and Shaw, B.D. (2010). Aspergillus nidulans 128,1465–1474. striatin (StrA) mediates sexual development and localizes to Simonin, A.R., Rasmussen, C.G., Yang, M., and Glass, N.L. (2010). the endoplasmic reticulum. Fungal Genet. Biol. 47, 789–799. Genes encoding a striatin-like protein (ham-3) and a forkhead Wei, L., Wang, Q., Zhang, Y., Yang, C., Guan, H., Chen, Y., and Sun, associated protein (ham-4) are required for hyphal fusion in Z. (2018). Identification of key genes, transcription factors and Neurospora crassa. Fungal Genet. Biol. 47, 855–868. involved in intracranial aneurysm. Mol. Med. Rep. Singh, N.S., Shao, N., McLean, J.R., Sevugan, M., Ren, L., Chew, T.G., 17, 891–897. Bimbo, A., Sharma, R., Tang, X., Gould, K.L., et al. (2011). SIN- Wong, M., Hyodo, T., Asano, E., Funasaka, K., Miyahara, R., Hirooka, inhibitory phosphatase complex promotes Cdc11p dephospho- Y., Goto, H., Hamaguchi, M., and Senga, T. (2014). Silencing rylation and propagates SIN asymmetry in fission yeast. Curr. of STRN 4 suppresses the malignant characteristics of cancer Biol. 21, 1968–1978. cells. Cancer Sci. 105, 1526–1532. Stegert, M.R., Hergovich, A., Tamaskovic, R., Bichsel, S.J., and Wu, S., Huang, J., Dong, J., and Pan, D. (2003). Hippo encodes a Hemmings, B.A. (2005). Regulation of NDR protein kinase by Ste-20 family protein kinase that restricts cell proliferation and hydrophobic motif phosphorylation mediated by the mamma- promotes apoptosis in conjunction with salvador and warts. lian Ste20-like kinase MST3. Mol. Cell. Biol. 25, 11019–11029. Cell 114, 445–456. Sung, V., Luo, W., Qian, D., Lee, I., Jallal, B., and Gishizky, M. (2003). Xiang, Q., Rasmussen, C., and Glass, N.L. (2002). The ham- The Ste20 kinase MST4 plays a role in prostate cancer progres- 2 locus, encoding a putative , is sion. Cancer Res. 63, 3356–3363. required for hyphal fusion in Neurospora crassa. Genetics Tang, F., Gill, J., Ficht, X., Barthlott, T., Cornils, H., Schmitz-Rohmer, 160, 169–180. D., Hynx, D., Zhou, D., Zhang, L., Xue, G., et al. (2015). The Xiong, W., Knox, A.J., Xu, M., Kiseljak-Vassiliades, K., Colgan, S.P., kinases NDR1/2 act downstream of the Hippo homolog MST1 Brodsky, K.S., Kleinschmidt-Demasters, B.K., Lillehei, K.O., to mediate both egress of thymocytes from the and and Wierman, M.E. (2015). Mammalian Ste20-like kinase 4 pro- lymphocyte motility. Sci. Signal 8, ra100. motes pituitary cell proliferation and survival under hypoxia. Tang, Y., Chen, M., Zhou, L., Ma, J., Li, Y., Zhang, H., Shi, Z., Xu, Mol. Endocrinol. 29, 460–472. Q., Zhang, X., Gao, Z., et al. (2019). Architecture, substruc- Zhang, H., Mukherjee, M., Kim, J.E., Yu, W., and Shim, W.B. (2018). tures, and dynamic assembly of STRIPAK complexes in Hippo Fsr1, a striatin homologue, forms an endomembrane-associ- ­signaling. Cell Discov. 5, 3. ated complex that regulates virulence in the maize pathogen Tanti, G.K. and Goswami, S.K. (2014). SG2NA recruits DJ-1 and Akt Fusarium verticillioides. Mol. Plant Pathol. 19, 812–826. into the mitochondria and membrane to protect cells from Zheng, Y., Liu, B., Wang, L., Lei, H., Pulgar Prieto, K.D., and Pan, D. oxidative damage Free Radic. Biol. Med. 75, 1–13. (2017). Homeostatic control of Hpo/MST kinase activity through Teichert, I., Nowrousian, M., Pöggeler, S., and Kück, U. (2014). autophosphorylation-dependent recruitment of the STRIPAK The filamentous fungus Sordaria macrospora as a genetic PP2A phosphatase complex. Cell Rep. 21, 3612–3623. model to study fruiting body development. Adv. Genet. 87, Zheng, S., Sun, P., Liu, H., Li, R., Long, L., Xu, Y., Chen, S., and Xu, J. 202–246. (2018). 17β-estradiol upregulates striatin protein levels via Akt Thompson, B.J. and Sahai, E. (2015). MST kinases in development pathway in human umbilical vein endothelial cells. PLoS One and disease. J. Cell Biol. 210, 871–882. 13, e0202500. 1022 U. Kück et al.: STRIPAK in development and disease

Ines Teichert Bionotes Allgemeine und Molekulare Botanik, Ruhr-Universität Bochum, Universitätsstr. Ulrich Kück 150, D-44780 Bochum, Germany Allgemeine und Molekulare Botanik, Ruhr-Universität Bochum, Universitätsstr. 150, D-44780 Bochum, Germany [email protected]

Ines Teichert leads an independent research group in the depart- ment of General and Molecular Botany and has a focus on develop- ment-dependent RNA editing. Ulrich Kück is a professor in the department of General and Molecu- lar Botany of the Ruhr University Bochum. He has a long-standing interest in the genetics of fungi.

Daria Radchenko Allgemeine und Molekulare Botanik, Ruhr-Universität Bochum, Universitätsstr. 150, D-44780 Bochum, Germany

Daria Radchenko is a postdoctoral fellow and has studied fungal signal transduction pathways, related to the Hippo signaling complex.