Next Article in Journal
Comparative Analysis of the GATA Transcription Factors in Five Solanaceae Species and Their Responses to Salt Stress in Wolfberry (Lycium barbarum L.)
Previous Article in Journal
Genomic Scanning of Inbreeding Depression for Litter Size in Two Varieties of Iberian Pigs
Previous Article in Special Issue
Exploring Large MAF Transcription Factors: Functions, Pathology, and Mouse Models with Point Mutations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Progress on Genetically Modified Animal Models for Membrane Skeletal Proteins: The 4.1 and MPP Families

1
Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
2
Center for Medical Education, Teikyo University of Science, Adachi-ku, Tokyo 120-0045, Japan
3
School of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan
4
Division of Basic & Clinical Medicine, Nagano College of Nursing, Komagane City, Nagano 399-4117, Japan
5
Division of Animal Research, Research Center for Advanced Science and Technology, Shinshu University, Matsumoto City, Nagano 390-8621, Japan
6
Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
*
Author to whom correspondence should be addressed.
Genes 2023, 14(10), 1942; https://doi.org/10.3390/genes14101942
Submission received: 2 October 2023 / Revised: 11 October 2023 / Accepted: 12 October 2023 / Published: 15 October 2023
(This article belongs to the Special Issue Animals Models in Diseases Genetics)

Abstract

:
The protein 4.1 and membrane palmitoylated protein (MPP) families were originally found as components in the erythrocyte membrane skeletal protein complex, which helps maintain the stability of erythrocyte membranes by linking intramembranous proteins and meshwork structures composed of actin and spectrin under the membranes. Recently, it has been recognized that cells and tissues ubiquitously use this membrane skeletal system. Various intramembranous proteins, including adhesion molecules, ion channels, and receptors, have been shown to interact with the 4.1 and MPP families, regulating cellular and tissue dynamics by binding to intracellular signal transduction proteins. In this review, we focus on our previous studies regarding genetically modified animal models, especially on 4.1G, MPP6, and MPP2, to describe their functional roles in the peripheral nervous system, the central nervous system, the testis, and bone formation. As the membrane skeletal proteins are located at sites that receive signals from outside the cell and transduce signals inside the cell, it is necessary to elucidate their molecular interrelationships, which may broaden the understanding of cell and tissue functions.

1. Protein 4.1 Family

1.1. Protein 4.1 in the Membrane Skeleton

Originally, membrane skeletal networks were found as a two-dimensional lattice structure beneath erythrocyte membranes, as schematically shown in Figure 1. Protein 4.1R–membrane palmitoylated protein 1 (MPP1)–glycophorin C is a basic molecular complex, in addition to ankyrin-band 3, attaching the actin–spectrin meshwork structures to form erythrocyte membrane skeletons, which support the erythrocyte membrane and provide stability, especially under blood flow [1]. Protein 4.1R (red cell) has 4.1–ezrin–radixin–moesin (FERM) and spectrin–actin binding (SAB) domains, and there are three other family members, namely 4.1B (brain), 4.1G (general), and 4.1N (nerve) [2,3]. In this review, we summarize recent studies on protein 4.1G in the peripheral nervous system (PNS) and bone development.

1.2. Protein 4.1G in PNS

Protein 4.1G was identified as FK506-binding protein 13 (FKBP13) [5]. We found its localization at two specific regions in Schwann cells that form myelin in the PNS: Schmidt–Lanterman incisures (SLIs) and paranodes [6]. Protein 4.1G assists in organizing internodes in the PNS [7], and is essential for the molecular targeting of MPP6 [8] and cell-adhesion molecule 4 (CADM4) [7] in SLIs. Thus, 4.1G–MPP6–CADM4, an analogous molecular complex to the erythrocyte membranes, exists in the PNS, likely functioning to resist external mechanical forces in SLIs [9]. 4.1G-deficient (-/-) mice showed motor impairment, especially with advancing age, and measurement of motor nerve velocity and the ultrastructure of myelin in the sciatic nerves demonstrated abnormalities under 4.1G-/- [10,11]. Considering that impairment of motor function with the tail-suspension test became worse after overwork treatment [11], careful attention is required in the rehabilitation of Charcot–Marie–Tooth (CMT) disease patients, which has been a controversial matter [12,13]. The SLI is thought to have function as a suspension structure against mechanical extension, similar to a spring [14], and in the case of 4.1G deficiency, the cell membrane may be destroyed.
CADM4 is probably related to the myelin abnormality under 4.1G-/- because the localization of CADM4 in SLIs disappears in 4.1G-/- nerves [15]. Furthermore, CADM4-/- nerves exhibited similar structural changes to those observed in human CMT disease [15,16]. CADM4 depletion and subsequent disruption may be related to erbB2 because they interact with each other [17,18]. Recent reports have shown that CADM1 has a role in maintaining cell–cell interspaces to promote the proper function of gap junction proteins [19,20]. Other than CADM4, several proteins, such as AP3 complex, tubulin, heat shock cognate 71 kD protein, and 14-3-3 protein, have been found that relate to 4.1G, from immunoprecipitation studies in the retina [21,22]. Because various proteins are associated with 4.1 families [2,23], it is necessary to further elucidate the binding proteins and functions for 4.1G in the PNS.
Additionally, it remains unclear how actin–spectrin components are connected to the 4.1G–MPP6–CADM4 complex in the PNS, considering that actin abundantly forms filaments in SLIs [24]. Notably, the SAB domain is spliced in the retina [22], and another actin-binding peptide sequence was found in 4.1R near the common SAB domain in epithelial cells [25]. Thus, the relationship between 4.1G and the actin filaments in SLIs has not been clarified.

1.3. Protein 4.1G in Bone Formation

Bone structure is controlled by the balance between bone formation by osteoblasts and bone resorption by osteoclasts. Osteoblasts are differentiated from mesenchymal stem cells and preosteoblasts (osteoblast differentiation). Many factors, including hedgehog, parathyroid hormone (PTH), and Wnt, affect osteoblast differentiation [26]. Moreover, 4.1G regulates hedgehog-mediated bone formation and PTH receptor (PTHR) signaling [27,28,29,30].
The primary cilium is a hair-like immotile sensory organelle that possesses selectively distributed membrane receptors, such as G-protein-coupled receptors (GPCRs) and growth factor receptors, and ion channels on its surrounding membrane (ciliary membrane) [31]. The cilium is formed in various cell types during the G0 phase of the cell cycle. A hedgehog receptor (i.e., smoothened) is one of the typical ciliary GPCRs expressed in the stem/progenitor cells of various organs (e.g., blood vessels, bone, brain, breast, esophagus, gallbladder, heart, intestine, liver, lung, pancreas, and stomach) [32,33,34,35]. Smoothened participates in the proliferation and differentiation of the cells to control organogenesis and tissue homeostasis.
Preosteoblasts form primary cilia on their surface. Deletion of the ciliary components, such as intraflagellar transport 80 (IFT80), IFT140, and kinesin 3a (Kif3a), disrupts preosteoblast ciliogenesis, ciliary hedgehog signaling, and femur or tibia formation [36,37,38]. Knockout of IFT20 in the cranial neural crest (CNC) disrupts ciliogenesis in CNC-derived osteogenic cells and leads to malformation of craniofacial bones [39]. These studies demonstrate the importance of primary cilia in bone formation. However, 4.1G is not recognized as a ciliary component, although it promotes ciliogenesis in preosteoblasts, as observed in the 4.1G-downregulated MC3T3-E1 preosteoblast cell line and 4.1G knockout preosteoblasts on trabecular bone in mouse new bone tibia [30]. In 4.1G-suppressed MC3T3-E1 cells, ciliary hedgehog signaling and subsequent osteoblast differentiation were attenuated, revealing a novel regulatory mechanism of bone formation by 4.1G.
Teriparatide, PTH-(1-34), is the first anabolic agent approved by the U.S. Food and Drug Administration for the treatment of osteoporosis [40]. Intermittent treatment with teriparatide facilitates osteoblast differentiation and suppresses osteoblast apoptosis [41,42]. Teriparatide activates PTHR, which is a GPCR. It strongly activates adenylyl cyclase (AC), produces cyclic AMP (cAMP) through Gs protein, and increases intracellular Ca2+ through Gq protein. In addition, 4.1G has been identified as an interacting protein of the carboxy (C)-terminus of PTHR [27]. Overexpression of 4.1G increases the amount of PTHR on the cell surface and PTHR-mediated intracellular Ca2+ elevation, suggesting that 4.1G augments the PTHR/Gq pathway by stabilizing the plasma membrane distribution of PTHR [27]. In contrast, PTHR/Gs-mediated cAMP production decreases with 4.1G overexpression and increases with 4.1G downregulation [28,29]. Mechanistically, 4.1G binds to the N-terminus of AC type 6 and attenuates its activity [29]. These studies suggest that 4.1G alters the signal balance of PTHR, with a high 4.1G expression, Gq > Gs, and with a low 4.1G expression, Gq < Gs. It is necessary to investigate whether the regulation of the PTHR signaling balance by 4.1G is one of the mechanisms in the intermittent treatment of teriparatide. Moreover, the ciliary distribution of PTHR and its role in bone formation have been identified; PTH-related protein treatment and shear stress stimuli promote translocation of PTHR to primary cilia, and the ciliary PTHR mediates cell survival and osteogenic gene expression in osteoblastic and osteoclastic cells [43,44,45]. The role of 4.1G in ciliary PTHR signaling remains unclarified.

2. MPP Family

2.1. MPP in Membrane Skeleton

In erythrocytes, the 4.1R–MPP1 (a.k.a. p55)–glycophorin C (GPC) molecular complex stabilizes erythrocyte membranes [46]. MPP1 belongs to the membrane-associated guanylate kinase homolog (MAGUK) family, which is characterized by the presence of the postsynaptic density protein 95 (PSD95)/Drosophila disc large tumor suppressor (Dlg)/zonula occludens 1 (ZO1) [PDZ] domain, Src-homology 3 (SH3) domain, and catalytic inactive guanylate kinase-like (GUK) domain [47]. The PDZ and SH3 domains can interact with lipids and proteins. The SH3 domain also has intramolecular and intermolecular interactions with the GUK domain. The GUK domain is thought to have low enzymatic activity, although the binding site for ATP and GMP in MPPs is intact. Except for MPP1, there are two L27 (Lin2- and Lin7-) domains, in which MPPs are capable of interacting with each other. Additionally, MPPs have a HOOK/D5 domain that binds to protein 4.1 members, and there are seven family members [48]. MPP1 binds to two distinct sites within the FERM domain of the 4.1 family, and the alternatively spliced exon 5 in 4.1R is necessary for the membrane targeting of 4.1R in epithelial cells [49]. In addition to the protein–protein interaction, palmitoylation helps transport MPP family proteins to cell membranes, and enzymes known as zinc finger DHHC-domain-containing palmitoyl acyl transferase (zDHHC/PATs) have roles in palmitoylation [50]. In this review, we summarize recent studies on MPP6 and MPP2 in the PNS, CNS, and testis.

2.2. MPP6 in PNS

As mentioned previously, 4.1G-/- mice showed that protein 4.1G is essential for the molecular targeting of MPP6 and CADM4 in SLIs in the PNS, as shown in Figure 2a [7,8,9]. We evaluated what would happen if MPP6 itself was deleted [51]. MPP6 deficiency also resulted in the hypermyelination of peripheral nerve fibers, although the phenotypes, such as structural changes and impairment of motor function, were weak compared with 4.1G deficiency.
The reason for hypermyelination without MPP6 was unclear. One of the MAGUK proteins, Dlg1 (SAP97), regulates membrane homeostasis in Schwann cells by interacting with kinesin 13B, Sec8, and myotubularin-related protein 2 (Mtmr2) for vesicle transport and membrane tethering [52]. The binding of the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) to the specific PDZ domain of Dlg1 inhibits axonal stimulation of myelination [53], and this Dlg1–PTEN complex is thought to limit myelin thickness to prevent overmyelination in the PNS [54]. Conditional inactivation of Dlg1 in Schwann cells caused a transient increase in myelin thickness during development, suggesting that Dlg1 is a transient regulator of myelination [55]. Deletion of the Dlg1–PTEN complex increases Akt phosphorylation and subsequent hypermyelination in peripheral nerves [56,57,58,59]. Additionally, disruption of PTEN in Schwann cells results in hyperactivation of the endogenous phosphoinositide 3-kinase (PI3K) pathway, focal hypermyelination, myelin outfoldings, and tomacula [60]. Dlg1 interacts with Mtmr2 [61] in phosphatidylinositol (PI) lipid metabolism [62]. These signals probably regulate the interaction between the actin cytoskeleton and plasma membrane interplay in a phosphoinositide cascade [63]. In addition, increased phosphatidylinositol (3,4,5)-triphosphate (PIP3) causes membrane wrapping and myelination [64]. In MPP1-deficient neutrophils, PIP3 forms punctate aggregations, which result in abnormal pseudopods [65]. Thus, our findings suggest that MPP6-deficient nerves may be related to the PTEN/Akt signal pathway.
The Src family of signal transduction proteins are also potentially related to the MPP family, because they interact with each other [66,67]. Additionally, as there are various PDZ-containing proteins in the PNS, such as MAGUK proteins (e.g., Dlg1 and MPP6), multi-PDZ domain protein 1 (MUPP1), pals-associated tight junction protein (PATJ), claudins, zonula occludens 1 (ZO1), and Par3 [68], but the extent to which they are interdependent or have mutual redundancy remains unclear.

2.3. MPPs and Lin7

2.3.1. Lin7 in PNS (Figure 2a)

Mammalian Lin7 (a.k.a. Veli/Mals) that contains L27 and PDZ domains was originally identified in a protein complex with the potential to couple synaptic vesicle exocytosis to cell adhesion in rat brains, and there are three family members [69]. Localization of Lin7 was found in SLIs, and MPP6 mainly transported Lin7 to SLIs in the mouse PNS [51]. Interactions between the Lin7 and MAGUK families have been reported in various tissues, including MPP4 recruitment of PSD95 and Lin7c (Veli3) in mouse photoreceptor synapses [70], MPP7 formation in a tripartite complex with Lin7 and Dlg1 in MDCK culture cells, which regulates the stability and localization of Dlg1 to cell junctions [71], and MPP4 and MPP5 association with Lin7c at distinct intercellular junctions of the mouse neurosensory retina [72]. The L27 domain is a scaffold for the supramolecular assembly of proteins in the Lin7 and MAGUK families [73,74,75]. Originally, both Pals family proteins, MPP5 (Pals1) and MPP6 (Pals2), were identified as proteins associated with Lin7 [76]. Although MPP5 was also reported in the PNS [77,78], our finding indicates that Lin7 transport in the PNS is mostly dependent on MPP6.

2.3.2. Lin7 in the CNS (Figure 2b)

In the cerebellum, high-resolution microscopic examination by Airy-confocal laser scanning microscopy revealed that the ring pattern in synaptic membrane staining and dot/spot areas inside synapses exhibited by Lin7 staining inversely correlated between MPP2+/+ and MPP2-/- synapses [79]. In MPP2-/- dendrites in cerebellar granular cells (GrCs), the Lin7-stained dot/spot areas did not overlap with the microtubule-associated protein 2 (MAP2)-stained dendritic shaft, indicating that MPP2 deficiency does not directly impair microtubule-based transport. In contrast, CADM1 exhibited a ring pattern in MPP2-/- synaptic membranes, and the number of Lin7-immunostained dot/spot areas localized inside the small CADM1-immunostained small rings was higher in MPP2-/- synapses than in MPP2+/+ ones. These results indicate MPP2 transports Lin7 from the dendritic shaft to postsynaptic membranes in synapses. Additionally, Lin7 was originally coimmunoprecipitated with CASK and Mint1, which bind to the vesicular trafficking protein Munc18-1 and are considered to play a role in the exocytosis of synaptic vesicles in presynaptic regions [69], whereas our findings demonstrated that Lin7 was abundantly localized at postsynaptic sites with MPP2 in GrCs in the cerebellum.

2.3.3. Lin7 in Testis (Figure 2c)

By immunohistochemistry (IHC), Lin7a and Lin7c were localized in germ cells, and Lin7c had especially strong staining in spermatogonia and early spermatocytes, characterized by staging of seminiferous tubules [80]. Lin7 staining became weaker in MPP6-/- testis according to both IHC and Western blotting, indicating a function of MPP6 in Lin7 transport in germ cells despite the unchanged histology of seminiferous tubules in MPP6-deficient mice compared with that of wild-type mice. In cultured spermatogonial stem cells maintained with glial-cell-line-derived neurotrophic factor, Lin7 was remarkably localized along cell membranes, especially at cell–cell junctions. Thus, Lin7 protein is localized in germ cells in relation to MPP6, which is a useful marker for spermatogenesis.

2.3.4. Proteins Interact with Lin7

Because MPP and protein 4.1 families are strongly related to Lin7 families, we listed the proteins associated with Lin7 from previous studies (Table 1) and categorized them into five groups. The first group is MAGUK family proteins and their relating proteins at cell–cell attaching sites, as described above in Section 2.3.1. The second group is the catenin–cadherin complex, an adhesion molecule. Aquaporin (AQP) 1 interacts with the Lin7–β-catenin complex in human melanoma and endothelial cell lines [81]. β-catenin and N-cadherin also interact with Lin7 in the rat brain [82], and the small GTPase Rho effector rhotekin interacts with the Lin7b–β-catenin complex in rat brain neurons [83]. In the third group, signal transduction proteins, such as the insulin receptor-substrate protein of 53 kD (IRSp53), are transported to tight junctions by Lin7 in cultured MDCK cells [84]. Signal transduction protein was detected at synapses in the rat cerebellum [85], and N-methyl-D-aspartate (NMDA) receptors increased in the IRSp53-knockout mouse hippocampus [86]. In the fourth group, synaptic proteins, such as GluN2B, bind to Lin7, and their complexes are carried by kinesin superfamily (KIF) 17 on microtubules in hippocampal neurons [87]. Interactions between the complex and PSD95 were also revealed in rat hippocampal postsynaptic regions [88].
In the fifth group, Lin7 interacts with several growth factor receptors. LET23 epidermal growth factor (EGF) receptor in Caenorhabditis elegans larval development [89] and Grindelwald tumor necrosis factor (TNF) receptor in Drosophila [90] are interesting examples, because they are related to the integration of cell signaling. Further examination of the Lin7 interaction with such receptors is necessary.
Concerning Lin7 knockout mice, although mice lacking Lin7a or Lin7c were viable and fertile, double knockout of mice for Lin7a and Lin7c was lethal before sexual maturation, suggesting that the functions of Lin7a and Lin7c likely compensated each other [91]. Additionally, Lin7a- and Lin7b-deficient mice are fertile and Lin7c was upregulated in mouse brain [92], indicating redundancy among Lin7 family members. Considering Lin7 in humans, disruption of cerebral cortex development by Lin7a depletion [93] and involvement in autism spectrum disorders by genetic alteration of Lin7b [94] has been reported. Therefore, target-cell-specific conditional disruption of Lin7 family proteins is required to elucidate the function of the Lin7 family.
Table 1. Associated proteins to Lin7 families.
Table 1. Associated proteins to Lin7 families.
Protein NameCategoryTissues and CellsMethodRelated ProteinsFunctional ConsiderationReferences
AQP12Human melanoma WM115 and endothelial HMEC1 cell linesIP, KDβ-cateninAQP1-KD affects Lin7/β-catenin expression[81]
BLT2 (Leukotriene B4 receptor)5MDCK cell linePD, KDCASK (Lin2)
Mint (Lin10)
Transportation from the Golgi apparatus to the plasma membrane[95]
BGT-1 (GABA transporter)4Recombinant Lin-7 and BGT-1 (PDZ target motif)BC Localization of transporter to plasma membranes[82]
CASK (Lin2)1Recombinant CASK, Velis proteins, rat brain
Mouse brain
IHC, YTH, IPMint (Lin10)Synaptic plasma membranes, synaptic vesicle exocytosis to cell adhesion[69]
CASK1Mouse brainBC, PDMint (Lin10)
KIF17
NR2B sorting vesicle carried by KIF17–Lin10 complex[87]
Crumbs (Drosophila)1Drosophila eye under Lin7 mutationIHC, PDStardust-PATJLight-dependent degeneration of photoreceptors[96]
β-catenin2Recombinant β-catenin and Lin7a, MDCK cell line and rat brain lysateBC, IPE-cadherinCadherin–β-catenin adhesion complex[82]
GluN2B (NMDA receptor)4Rat cerebral cortex, transfected NR2B or MALSIP, PDPSD95MALS2 directly binds to NR2B[88]
Grindelwald (Drosophila; TNF receptor)5Transfection of mutated Lin7IHCStardust-PATJ-CrumbsTransport of TNF (tumor necrosis factor) receptor[90]
IRSp533Rat brain, MDCK cell lineYTH, IPSAP102Formation or maintenance of the adhesion structure of epithelium[97]
LET-23 (C. elegans; EGF receptor)5Transfection of mutated Lin-7IHC, YTHCASK (Lin2) Vulval induction[98]
LET-235Transfection of mutated Lin-7IHCLin2-Lin10 complexTransport of LET-23 from the Golgi apparatus to the cell membrane[89]
Mint (Lin10)1Rat homolog of the C. elegans Lin10Cloning, IHCCASK (Lin2) Distributed in the membrane fraction in rat brain [99]
MPP41Porcine retinal membranes
Transfection of bovine MPP4 L27C or L27N + C domain
IP,
PD
MPP5Veli3 and MPP4 most intense staining in photoreceptor terminals of the outer plexiform layer (OPL)[72]
MPP5 (Pals1)1Cloning of Lin-7 binding partnersPDMPP6
CASK (Lin2)
Localize to the lateral membrane[76]
MPP6 (VAM1, Pals2)1Cloning of Lin7 binding partnersCloning, PDMPP5
CASK (Lin2)
Localize to the lateral membrane[76]
MPP61Transfection of human Veli1 binds to VAM1PD MPP6 does not bind to 4.1R
[100]
MPP71Transfected human
MPP7 L27C domain
PDDlg1Enhanced localization of Dlg1 to cell junction[71]
Rhotekin2COS7 cells and rat brainYTHPISTTrafficking of protein in synapses[101]
Stardust (Drosophila; Pals1)1Transfection of mutated Lin7IHCCrumbsTransport of Grindelwalt (homologous to TNFR)[90]
Category: Lin7-associating proteins are categorized into five groups as described in the text. BC: biochemical binding assay, IHC: immunohistochemistry, IP: immunoprecipitation, PD: pull down, YTH: yeast two-hybrid system.

2.4. MPPs and CADMs

CADMs are Ca2+-independent adhesion molecules, and they have binding properties to both protein 4.1 and MPPs [102]. In the PNS, deficiency of the MPP6–Lin7 complex had little effect on CADM4, and cadherin and tight-junction proteins were retained [51]. However, scaffolding for CADM4 in SLI is mostly dependent on protein 4.1G, as shown in Figure 2a [15,16,51]. In testes, the expression and localization of CADM1 were retained in 4.1G/4.1B double-/- and MPP6-/- mice, as shown in Figure 2c [8,10,80].
In the CNS, scaffolding for CADMs is more complicated, because many MAGUKs are associated with CADM1 [103,104]. Although the PDZ domain of MPP2 was reported to directly interact with the C-terminus of CADM1 in rat hippocampal neurons [105], and nearly 80% of MPP2 dots overlapped with CADM1 areas by IHC and cerebellar lysate of MPP2 included CADM1 by immunoprecipitation study in our recent study in cerebellum, MPP2-/- synapses did not show reduction of CADM1 in cerebellar GrCs, as shown in Figure 2b [79]. Considering that CADM1-/- mice exhibited small cerebella with a decreased number of synapses compared with wild-type mice [106], the redundancy of MAGUK and 4.1 families to locate CADM family proteins has not been clarified.

2.5. MPP and Neurotransmitters

MPP2 specifically localizes to the cerebellar granular layer, particularly to dendritic terminals in GrCs facing the mossy fiber (MF) terminus at the cerebellar glomerulus, as schematically summarized with MPP2-interactive proteins in Figure 3a [79], because the MF–GrC synapses are the first place to transduce excitatory electrical signals into cerebellum [107]. MAGUK family proteins, such as PSD95 (Dlg4, SAP90), SAP102 (Dlg3), and Chapsyn-110 (Dlg2, PSD93), localize to both the molecular and granular layers [108]. To clarify the specific localization of MPP2, localizations of various MAGUKs are demonstrated in Figure 3b–k. Note that the gene loci of MPP2 (in mouse chromosome 11) and Dlg2 (in mouse chromosome 7) are different.
MAGUKs are known to associate with excitatory NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) glutamate neurotransmitters [109,110]. At MF–GrC synapses, NMDA receptors such as GluN1 and GluN2A/C were detected [111] as well as adherens junctions consisting of GrC dendrites [112]. Additionally, transmembrane AMPA receptor regulatory proteins (TARPs) γ2/γ7 were detected in the postsynaptic regions of MF–GrC synapses with AMPA receptors GluA2/GluA4 [113]. GrC-specific GluA4-knockout mice showed a delay in eyeblink conditioning, but not locomotor coordination [114]. Motor dysfunction with a simple walking test has not been detected in MPP2-/- mice [79], and further examination for the neurological examination under conditioning is necessary.
In addition to excitatory neurotransmitters, a recent study demonstrated that MPP2 interacts with inhibitory γ-amino butyric acid (GABA) neurotransmitters without the involvement of gephyrin in rat hippocampal neurons [115]. In the cerebellar glomerulus, GABAergic neurotransmission is mediated between Golgi cells and GrCs, and two types of GABAergic inhibition have been proposed: phasic and tonic inhibition [116]. For phasic inhibition (transient inhibition), GABAAR consists of α1, α6, β2/3, and γ2 subunits in synaptic regions, and, for tonic inhibition (sustained inhibition), GABAAR consists of α1, α6, β2/3, and δ2 subunits in extrasynaptic regions [116]. GABAARs in synaptic regions interact with neuroligin 2, GABAAR regulatory Lhfpl, gephyrin [117], and synaptic scaffolding molecule (S-SCAM)/membrane associated guanylate kinase 2 (MAGI2) [118].
Figure 4 shows double-immunostaining and Airyscan-confocal laser scanning microscopy observations, demonstrating comparative localizations of MPP2 to α1, gephyrin, and α6. Gephyrin is a scaffold protein in the synaptic region, and α6 is a GABAAR in the extrasynaptic region. α1 (Figure 4a,c,f,h,k,m) and α6 (Figure 4l,m) staining were observed as dot/line patterns, whereas MPP2 (Figure 4b,c) and gephyrin (Figure 4g,h) staining were recognized as dot patterns in CG. Approximately 44% of the α1-stained areas (n = 94) overlapped with the MPP2-stained dots (Figure 4e), indicating a relationship between α1 and MPP2. Additionally, ~43% of the gephyrin-stained dots (n = 65) overlapped with α1-stained areas (Figure 4j), and ~27% of the α1-stained areas (n = 74) overlapped with α6-stained areas (Figure 4o). Thus, the overlap of the α1/MPP2 areas with the gephyrin and α6 areas indicate that α1/MPP2 localize in synaptic and extrasynaptic regions, respectively.
As MPP2 was reported to interact with several GABAAR subunits [115] and various subunits are present in the cerebellum [119], it is necessary to consider the interdependence of the GABAAR subunits. In the thalamus of the α4-knockout mouse, δ was decreased, whereas α1 and γ2 were increased in extrasynaptic regions, suggesting compensation among GABAAR subunits [120]. In addition, in the α1-knockout mouse, increases in the α3, α4, and α6 subunits, reductions in the β2/3 and γ2 subunits, and maintenance of the α5 and δ subunits were reported [121]. Further studies on the balance of these GABAAR subunits under MPP deficiency are necessary.
Several membrane skeletal proteins have been reported to interact with GABAAR. A giant ankyrin-G controls endocytosis of GABAAR by interacting with GABAAR-associated protein (GABARAP) in the mouse-cultured hippocampus [122]. GABAARα5 interacts with a membrane skeletal ezrin–radixin–moesin family protein, radixin, in mouse hippocampus [123]. GABAAR also interacts with neuroligin1 and CASK in inhibitory neuromuscular junctions in C. elegans [124]. MPP2 may be dependent on these membrane skeletal proteins to locate GABAAR.

2.6. MPP Families in Synapses

MAGUK proteins become oligomers because of PDZ–SH3–GUK tandem domains, function as a molecular complex in cell membranes specifically at cell–cell adhesion areas, and occur in various tissues and organs [125,126]. Particularly, there are many MAGUK family proteins in synapses, which function in postsynaptic density formation and signal transduction, and their impairment is related to some mental diseases [110,127,128,129,130]. A recent genome-wide association study (GWAS) also demonstrated the relationship between MPP6 and various psychiatric disorders: the MPP6 gene was included in 64 genome loci for bipolar disorders compared among European ancestry [131], in 109 genome loci associated with at least two psychiatric disorders including anorexia nervosa, attention-deficit/hyperactivity disorder, major depression, obsessive–compulsive disorder, schizophrenia, and Tourette syndrome [132], and in 108 genome loci for schizophrenia patients [133]. MPP6 was also included in 57 hard sweep genes after the initial movement of the evolutionarily recent dispersal of anatomically modern humans out of Africa, among genes related to biological processes, including ciliopathies, metabolic syndrome, and neurodegenerative disorders. [134]. In addition, a GWAS for sleep disorders demonstrated novel genome-wide loci on human chromosome 7 between NPY and MPP6, and disruption of an ortholog of MPP6 in Drosophila melanogaster was identified in sleep center neurons relating to decreased sleep duration [135]. In these respects, it is necessary to evaluate neurological psychological impairments in genetically modified MPP-deficient mice, which may be related to human diseases that are caused by mutation in MPP genes.

3. Conclusions

The 4.1 and MPP families are not only membrane skeletal components but are also widely distributed in various organs to transport intramembranous and signal transduction proteins. Especially, 4.1G has an obvious function in myelin formation in the PNS. There may be some interdependence and redundancy among the 4.1 and MPP families, as well as related proteins in other organs such as the CNS and testis, which brings about future challenges to examining cross-breeds of several genetically modified model mice. Considering that the molecular evolution of vertebrate behaviors may be related to the diversity of MAGUK proteins including MPPs [136], further evaluation of a wide range of molecular complexes, by proteomic and transcriptome analyses combined with genetically modified animal models, may broaden the understanding of normal morphological and physiological functions as well as physical and mental impairment.

Author Contributions

N.T., Y.S., M.S., T.Y. (Tomoki Yamada), A.K., T.Y. (Takahiro Yoshizawa) and T.S. performed the experiments, analyzed the data, and wrote the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by grants from the Japan Society for the Promotion of Science, KAKENHI 23K10424 to N.T. and KAKENHI 21K11275 to Y.S.

Institutional Review Board Statement

The animal study protocol was approved by the Institutional Review Board of Shinshu University.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lux, S.E., IV. Anatomy of the red cell membrane skeleton: Unanswered questions. Blood 2016, 127, 187–199. [Google Scholar] [CrossRef] [PubMed]
  2. Baines, A.J.; Lu, H.C.; Bennett, P.M. The Protein 4.1 family: Hub proteins in animals for organizing membrane proteins. Biochim. Biophys. Acta 2014, 1838, 605–619. [Google Scholar] [CrossRef]
  3. Baines, A.J. Link up and fold up-templating the formation of spectrin tetramers. J. Mol. Biol. 2014, 426, 7–10. [Google Scholar] [CrossRef]
  4. Lux, S.E.; Wolfe, L.C.; Pease, B.; Tomaselli, M.B.; John, K.M.; Bernstein, S.E. Hemolytic anemias due to abnormalities in red cell spectrin: A brief review. Prog. Clin. Biol. Res. 1981, 45, 159–168. [Google Scholar]
  5. Walensky, L.D.; Gascard, P.; Fields, M.E.; Blackshaw, S.; Conboy, J.G.; Mohandas, N.; Snyder, S.H. The 13-kD FK506 binding protein, FKBP13, interacts with a novel homologue of the erythrocyte membrane cytoskeletal protein 4.1. J. Cell Biol. 1998, 141, 143–153. [Google Scholar] [CrossRef] [PubMed]
  6. Ohno, N.; Terada, N.; Yamakawa, H.; Komada, M.; Ohara, O.; Trapp, B.D.; Ohno, S. Expression of protein 4.1G in Schwann cells of the peripheral nervous system. J. Neurosci. Res. 2006, 84, 568–577. [Google Scholar] [CrossRef] [PubMed]
  7. Ivanovic, A.; Horresh, I.; Golan, N.; Spiegel, I.; Sabanay, H.; Frechter, S.; Ohno, S.; Terada, N.; Mobius, W.; Rosenbluth, J.; et al. The cytoskeletal adapter protein 4.1G organizes the internodes in peripheral myelinated nerves. J. Cell Biol. 2012, 196, 337–344. [Google Scholar] [CrossRef] [PubMed]
  8. Terada, N.; Saitoh, Y.; Ohno, N.; Komada, M.; Saitoh, S.; Peles, E.; Ohno, S. Essential function of protein 4.1G in targeting of membrane protein palmitoylated 6 into Schmidt-Lanterman incisures in myelinated nerves. Mol. Cell Biol. 2012, 32, 199–205. [Google Scholar] [CrossRef] [PubMed]
  9. Terada, N.; Saitoh, Y.; Kamijo, A.; Ohno, S.; Ohno, N. Involvement of membrane skeletal molecules in the Schmidt-Lanterman incisure in Schwann cells. Med. Mol. Morphol. 2016, 49, 5–10. [Google Scholar] [CrossRef]
  10. Terada, N.; Ohno, N.; Saitoh, S.; Saitoh, Y.; Komada, M.; Kubota, H.; Ohno, S. Involvement of a membrane skeletal protein, 4.1G, for Sertoli/germ cell interaction. Reproduction 2010, 139, 883–892. [Google Scholar] [CrossRef] [PubMed]
  11. Saitoh, Y.; Ohno, N.; Yamauchi, J.; Sakamoto, T.; Terada, N. Deficiency of a membrane skeletal protein, 4.1G, results in myelin abnormalities in the peripheral nervous system. Histochem. Cell Biol. 2017, 148, 597–606. [Google Scholar] [CrossRef]
  12. Van Pomeren, M.; Selles, R.W.; van Ginneken, B.T.; Schreuders, T.A.; Janssen, W.G.; Stam, H.J. The hypothesis of overwork weakness in Charcot-Marie-Tooth: A critical evaluation. J. Rehabil. Med. 2009, 41, 32–34. [Google Scholar] [CrossRef]
  13. Vinci, P.; Perelli, S.L.; Gargiulo, P. About the hypothesis of overwork weakness in Charcot-Marie-Tooth disease. J. Rehabil. Med. 2009, 41, 778. [Google Scholar] [CrossRef]
  14. Kamijo, A.; Saitoh, Y.; Ohno, N.; Ohno, S.; Terada, N. Immunohistochemical study of mouse sciatic nerves under various stretching conditions with “in vivo cryotechnique”. J. Neurosci. Methods 2014, 227, 181–188. [Google Scholar] [CrossRef] [PubMed]
  15. Golan, N.; Kartvelishvily, E.; Spiegel, I.; Salomon, D.; Sabanay, H.; Rechav, K.; Vainshtein, A.; Frechter, S.; Maik-Rachline, G.; Eshed-Eisenbach, Y.; et al. Genetic deletion of Cadm4 results in myelin abnormalities resembling Charcot-Marie-Tooth neuropathy. J. Neurosci. 2013, 33, 10950–10961. [Google Scholar] [CrossRef]
  16. Spiegel, I.; Adamsky, K.; Eshed, Y.; Milo, R.; Sabanay, H.; Sarig-Nadir, O.; Horresh, I.; Scherer, S.S.; Rasband, M.N.; Peles, E. A central role for Necl4 (SynCAM4) in Schwann cell-axon interaction and myelination. Nat. Neurosci. 2007, 10, 861–869. [Google Scholar] [CrossRef] [PubMed]
  17. Kawano, S.; Ikeda, W.; Kishimoto, M.; Ogita, H.; Takai, Y. Silencing of ErbB3/ErbB2 signaling by immunoglobulin-like Necl-2. J. Biol. Chem. 2009, 284, 23793–23805. [Google Scholar] [CrossRef] [PubMed]
  18. Mizutani, K.; Kedashiro, S.; Maruoka, M.; Ueda, Y.; Takai, Y. Nectin-like molecule-4/cell adhesion molecule 4 inhibits the ligand-induced dimerization of ErbB3 with ErbB2. Sci. Rep. 2017, 7, 11375. [Google Scholar] [CrossRef] [PubMed]
  19. Wu, X.; Azizan, E.A.B.; Goodchild, E.; Garg, S.; Hagiyama, M.; Cabrera, C.P.; Fernandes-Rosa, F.L.; Boulkroun, S.; Kuan, J.L.; Tiang, Z.; et al. Somatic mutations of CADM1 in aldosterone-producing adenomas and gap junction-dependent regulation of aldosterone production. Nat. Genet. 2023, 55, 1009–1021. [Google Scholar] [CrossRef]
  20. Ito, A.; Ichiyanagi, N.; Ikeda, Y.; Hagiyama, M.; Inoue, T.; Kimura, K.B.; Sakurai, M.A.; Hamaguchi, K.; Murakami, Y. Adhesion molecule CADM1 contributes to gap junctional communication among pancreatic islet α-cells and prevents their excessive secretion of glucagon. Islets 2012, 4, 49–55. [Google Scholar] [CrossRef]
  21. Cheng, C.L.; Molday, R.S. Interaction of 4.1G and cGMP-gated channels in rod photoreceptor outer segments. J. Cell Sci. 2013, 126, 5725–5734. [Google Scholar] [CrossRef]
  22. Sanuki, R.; Watanabe, S.; Sugita, Y.; Irie, S.; Kozuka, T.; Shimada, M.; Ueno, S.; Usukura, J.; Furukawa, T. Protein-4.1G-mediated membrane trafficking is essential for correct rod synaptic location in the retina and for normal visual function. Cell Rep. 2015, 10, 796–808. [Google Scholar] [CrossRef] [PubMed]
  23. Yang, Q.; Liu, J.; Wang, Z. 4.1N-mediated interactions and functions in nerve system and cancer. Front. Mol. Biosci. 2021, 8, 711302. [Google Scholar] [CrossRef] [PubMed]
  24. Trapp, B.D.; Andrews, S.B.; Wong, A.; O’Connell, M.; Griffin, J.W. Co-localization of the myelin-associated glycoprotein and the microfilament components, F-actin and spectrin, in Schwann cells of myelinated nerve fibres. J. Neurocytol. 1989, 18, 47–60. [Google Scholar] [CrossRef] [PubMed]
  25. Huang, S.C.; Liang, J.Y.; Vu, L.V.; Yu, F.H.; Ou, A.C.; Ou, J.P.; Zhang, H.S.; Burnett, K.M.; Benz, E.J., Jr. Epithelial-specific isoforms of protein 4.1R promote adherens junction assembly in maturing epithelia. J. Biol. Chem. 2020, 295, 191–211. [Google Scholar] [CrossRef]
  26. Rutkovskiy, A.; Stenslokken, K.O.; Vaage, I.J. Osteoblast differentiation at a glance. Med. Sci. Monit. Basic Res. 2016, 22, 95–106. [Google Scholar] [CrossRef]
  27. Saito, M.; Sugai, M.; Katsushima, Y.; Yanagisawa, T.; Sukegawa, J.; Nakahata, N. Increase in cell-surface localization of parathyroid hormone receptor by cytoskeletal protein 4.1G. Biochem. J. 2005, 392, 75–81. [Google Scholar] [CrossRef] [PubMed]
  28. Goto, T.; Chiba, A.; Sukegawa, J.; Yanagisawa, T.; Saito, M.; Nakahata, N. Suppression of adenylyl cyclase-mediated cAMP production by plasma membrane associated cytoskeletal protein 4.1G. Cell Signal. 2013, 25, 690–697. [Google Scholar] [CrossRef] [PubMed]
  29. Saito, M.; Cui, L.; Hirano, M.; Li, G.; Yanagisawa, T.; Sato, T.; Sukegawa, J. Activity of adenylyl cyclase type 6 is suppressed by direct binding of the cytoskeletal protein 4.1G. Mol. Pharmacol. 2019, 96, 441–451. [Google Scholar] [CrossRef]
  30. Saito, M.; Hirano, M.; Izumi, T.; Mori, Y.; Ito, K.; Saitoh, Y.; Terada, N.; Sato, T.; Sukegawa, J. Cytoskeletal Protein 4.1G is essential for the primary ciliogenesis and osteoblast differentiation in bone formation. Int. J. Mol. Sci. 2022, 23, 2094. [Google Scholar] [CrossRef]
  31. Pala, R.; Alomari, N.; Nauli, S.M. Primary cilium-dependent signaling mechanisms. Int. J. Mol. Sci. 2017, 18, 2272. [Google Scholar] [CrossRef] [PubMed]
  32. Echelard, Y.; Epstein, D.J.; St-Jacques, B.; Shen, L.; Mohler, J.; McMahon, J.A.; McMahon, A.P. Sonic hedgehog, a member of a family of putative signaling molecules, is implicated in the regulation of CNS polarity. Cell 1993, 75, 1417–1430. [Google Scholar] [CrossRef] [PubMed]
  33. Briscoe, J.; Therond, P.P. The mechanisms of Hedgehog signalling and its roles in development and disease. Nat. Rev. Mol. Cell Biol. 2013, 14, 416–429. [Google Scholar] [CrossRef] [PubMed]
  34. Jeng, K.S.; Chang, C.F.; Lin, S.S. Sonic hedgehog signaling in organogenesis, tumors, and tumor microenvironments. Int. J. Mol. Sci. 2020, 21, 758. [Google Scholar] [CrossRef] [PubMed]
  35. Shimada, I.S.; Kato, Y. Ciliary signaling in stem cells in health and disease: Hedgehog pathway and beyond. Semin. Cell Dev. Biol. 2022, 129, 115–125. [Google Scholar] [CrossRef]
  36. Qiu, N.; Xiao, Z.; Cao, L.; Buechel, M.M.; David, V.; Roan, E.; Quarles, L.D. Disruption of Kif3a in osteoblasts results in defective bone formation and osteopenia. J. Cell Sci. 2012, 125, 1945–1957. [Google Scholar] [CrossRef]
  37. Yuan, X.; Cao, J.; He, X.; Serra, R.; Qu, J.; Cao, X.; Yang, S. Ciliary IFT80 balances canonical versus non-canonical hedgehog signalling for osteoblast differentiation. Nat. Commun. 2016, 7, 11024. [Google Scholar] [CrossRef] [PubMed]
  38. Chen, Y.; Fan, Q.; Zhang, H.; Tao, D.; Wang, Y.; Yue, R.; Sun, Y. Lineage tracing of cells expressing the ciliary gene IFT140 during bone development. Dev. Dyn. 2021, 250, 574–583. [Google Scholar] [CrossRef]
  39. Noda, K.; Kitami, M.; Kitami, K.; Kaku, M.; Komatsu, Y. Canonical and noncanonical intraflagellar transport regulates craniofacial skeletal development. Proc. Natl. Acad. Sci. USA 2016, 113, E2589–E2597. [Google Scholar] [CrossRef]
  40. Uihlein, A.V.; Leder, B.Z. Anabolic therapies for osteoporosis. Endocrinol. Metab. Clin. N. Am. 2012, 41, 507–525. [Google Scholar] [CrossRef]
  41. Jilka, R.L.; Weinstein, R.S.; Bellido, T.; Roberson, P.; Parfitt, A.M.; Manolagas, S.C. Increased bone formation by prevention of osteoblast apoptosis with parathyroid hormone. J. Clin. Investig. 1999, 104, 439–446. [Google Scholar] [CrossRef]
  42. Balani, D.H.; Ono, N.; Kronenberg, H.M. Parathyroid hormone regulates fates of murine osteoblast precursors in vivo. J. Clin. Investig. 2017, 127, 3327–3338. [Google Scholar] [CrossRef]
  43. Zheng, L.; Cao, Y.; Ni, S.; Qi, H.; Ling, Z.; Xu, X.; Zou, X.; Wu, T.; Deng, R.; Hu, B.; et al. Ciliary parathyroid hormone signaling activates transforming growth factor-β to maintain intervertebral disc homeostasis during aging. Bone Res. 2018, 6, 21. [Google Scholar] [CrossRef]
  44. Martin-Guerrero, E.; Tirado-Cabrera, I.; Buendia, I.; Alonso, V.; Gortazar, A.R.; Ardura, J.A. Primary cilia mediate parathyroid hormone receptor type 1 osteogenic actions in osteocytes and osteoblasts via Gli activation. J. Cell Physiol. 2020, 235, 7356–7369. [Google Scholar] [CrossRef] [PubMed]
  45. Tirado-Cabrera, I.; Martin-Guerrero, E.; Heredero-Jimenez, S.; Ardura, J.A.; Gortazar, A.R. PTH1R translocation to primary cilia in mechanically-stimulated ostecytes prevents osteoclast formation via regulation of CXCL5 and IL-6 secretion. J. Cell Physiol. 2022, 237, 3927–3943. [Google Scholar] [CrossRef]
  46. Nunomura, W.; Takakuwa, Y.; Parra, M.; Conboy, J.; Mohandas, N. Regulation of protein 4.1R, p55, and glycophorin C ternary complex in human erythrocyte membrane. J. Biol. Chem. 2000, 275, 24540–24546. [Google Scholar] [CrossRef]
  47. Dimitratos, S.D.; Woods, D.F.; Stathakis, D.G.; Bryant, P.J. Signaling pathways are focused at specialized regions of the plasma membrane by scaffolding proteins of the MAGUK family. BioEssays News Rev. Mol. Cell. Dev. Biol. 1999, 21, 912–921. [Google Scholar] [CrossRef]
  48. Chytla, A.; Gajdzik-Nowak, W.; Olszewska, P.; Biernatowska, A.; Sikorski, A.F.; Czogalla, A. Not just another scaffolding protein family: The multifaceted MPPs. Molecules 2020, 25, 4954. [Google Scholar] [CrossRef] [PubMed]
  49. Seo, P.S.; Jeong, J.J.; Zeng, L.; Takoudis, C.G.; Quinn, B.J.; Khan, A.A.; Hanada, T.; Chishti, A.H. Alternatively spliced exon 5 of the FERM domain of protein 4.1R encodes a novel binding site for erythrocyte p55 and is critical for membrane targeting in epithelial cells. Biochim. Biophys. Acta 2009, 1793, 281–289. [Google Scholar] [CrossRef]
  50. Fukata, Y.; Fukata, M. Protein palmitoylation in neuronal development and synaptic plasticity. Nat. Rev. Neurosci. 2010, 11, 161–175. [Google Scholar] [CrossRef] [PubMed]
  51. Saitoh, Y.; Kamijo, A.; Yamauchi, J.; Sakamoto, T.; Terada, N. The membrane palmitoylated protein, MPP6, is involved in myelin formation in the mouse peripheral nervous system. Histochem. Cell Biol. 2019, 151, 385–394. [Google Scholar] [CrossRef] [PubMed]
  52. Bolis, A.; Coviello, S.; Visigalli, I.; Taveggia, C.; Bachi, A.; Chishti, A.H.; Hanada, T.; Quattrini, A.; Previtali, S.C.; Biffi, A.; et al. Dlg1, Sec8, and Mtmr2 regulate membrane homeostasis in Schwann cell myelination. J. Neurosci. 2009, 29, 8858–8870. [Google Scholar] [CrossRef] [PubMed]
  53. Valiente, M.; Andres-Pons, A.; Gomar, B.; Torres, J.; Gil, A.; Tapparel, C.; Antonarakis, S.E.; Pulido, R. Binding of PTEN to specific PDZ domains contributes to PTEN protein stability and phosphorylation by microtubule-associated serine/threonine kinases. J. Biol. Chem. 2005, 280, 28936–28943. [Google Scholar] [CrossRef]
  54. Cotter, L.; Ozcelik, M.; Jacob, C.; Pereira, J.A.; Locher, V.; Baumann, R.; Relvas, J.B.; Suter, U.; Tricaud, N. Dlg1-PTEN interaction regulates myelin thickness to prevent damaging peripheral nerve overmyelination. Science 2010, 328, 1415–1418. [Google Scholar] [CrossRef]
  55. Noseda, R.; Belin, S.; Piguet, F.; Vaccari, I.; Scarlino, S.; Brambilla, P.; Martinelli Boneschi, F.; Feltri, M.L.; Wrabetz, L.; Quattrini, A.; et al. DDIT4/REDD1/RTP801 is a novel negative regulator of Schwann cell myelination. J. Neurosci. 2013, 33, 15295–15305. [Google Scholar] [CrossRef]
  56. Domenech-Estevez, E.; Baloui, H.; Meng, X.; Zhang, Y.; Deinhardt, K.; Dupree, J.L.; Einheber, S.; Chrast, R.; Salzer, J.L. Akt regulates axon wrapping and myelin sheath thickness in the PNS. J. Neurosci. 2016, 36, 4506–4521. [Google Scholar] [CrossRef] [PubMed]
  57. Figlia, G.; Gerber, D.; Suter, U. Myelination and mTOR. Glia 2018, 66, 693–707. [Google Scholar] [CrossRef]
  58. Norrmen, C.; Suter, U. Akt/mTOR signalling in myelination. Biochem. Soc. Trans. 2013, 41, 944–950. [Google Scholar] [CrossRef]
  59. Pereira, J.A.; Lebrun-Julien, F.; Suter, U. Molecular mechanisms regulating myelination in the peripheral nervous system. Trends Neurosci. 2012, 35, 123–134. [Google Scholar] [CrossRef] [PubMed]
  60. Goebbels, S.; Oltrogge, J.H.; Wolfer, S.; Wieser, G.L.; Nientiedt, T.; Pieper, A.; Ruhwedel, T.; Groszer, M.; Sereda, M.W.; Nave, K.A. Genetic disruption of Pten in a novel mouse model of tomaculous neuropathy. EMBO Mol. Med. 2012, 4, 486–499. [Google Scholar] [CrossRef]
  61. Bolino, A.; Bolis, A.; Previtali, S.C.; Dina, G.; Bussini, S.; Dati, G.; Amadio, S.; Del Carro, U.; Mruk, D.D.; Feltri, M.L.; et al. Disruption of Mtmr2 produces CMT4B1-like neuropathy with myelin outfolding and impaired spermatogenesis. J. Cell Biol. 2004, 167, 711–721. [Google Scholar] [CrossRef]
  62. Guerrero-Valero, M.; Grandi, F.; Cipriani, S.; Alberizzi, V.; Di Guardo, R.; Chicanne, G.; Sawade, L.; Bianchi, F.; Del Carro, U.; De Curtis, I.; et al. Dysregulation of myelin synthesis and actomyosin function underlies aberrant myelin in CMT4B1 neuropathy. Proc. Natl. Acad. Sci. USA 2021, 118, e2009469118. [Google Scholar] [CrossRef] [PubMed]
  63. Saarikangas, J.; Zhao, H.; Lappalainen, P. Regulation of the actin cytoskeleton-plasma membrane interplay by phosphoinositides. Physiol. Rev. 2010, 90, 259–289. [Google Scholar] [CrossRef]
  64. Goebbels, S.; Oltrogge, J.H.; Kemper, R.; Heilmann, I.; Bormuth, I.; Wolfer, S.; Wichert, S.P.; Mobius, W.; Liu, X.; Lappe-Siefke, C.; et al. Elevated phosphatidylinositol 3,4,5-trisphosphate in glia triggers cell-autonomous membrane wrapping and myelination. J. Neurosci. 2010, 30, 8953–8964. [Google Scholar] [CrossRef]
  65. Quinn, B.J.; Welch, E.J.; Kim, A.C.; Lokuta, M.A.; Huttenlocher, A.; Khan, A.A.; Kuchay, S.M.; Chishti, A.H. Erythrocyte scaffolding protein p55/MPP1 functions as an essential regulator of neutrophil polarity. Proc. Natl. Acad. Sci. USA 2009, 106, 19842–19847. [Google Scholar] [CrossRef]
  66. Baumgartner, M.; Weiss, A.; Fritzius, T.; Heinrich, J.; Moelling, K. The PDZ protein MPP2 interacts with c-Src in epithelial cells. Exp. Cell Res. 2009, 315, 2888–2898. [Google Scholar] [CrossRef] [PubMed]
  67. Terada, N.; Saitoh, Y.; Ohno, N.; Komada, M.; Yamauchi, J.; Ohno, S. Involvement of Src in the membrane skeletal complex, MPP6-4.1G, in Schmidt-Lanterman incisures of mouse myelinated nerve fibers in PNS. Histochem. Cell Biol. 2013, 140, 213–222. [Google Scholar] [CrossRef] [PubMed]
  68. Poliak, S.; Matlis, S.; Ullmer, C.; Scherer, S.S.; Peles, E. Distinct claudins and associated PDZ proteins form different autotypic tight junctions in myelinating Schwann cells. J. Cell Biol. 2002, 159, 361–372. [Google Scholar] [CrossRef] [PubMed]
  69. Butz, S.; Okamoto, M.; Sudhof, T.C. A tripartite protein complex with the potential to couple synaptic vesicle exocytosis to cell adhesion in brain. Cell 1998, 94, 773–782. [Google Scholar] [CrossRef]
  70. Aartsen, W.M.; Kantardzhieva, A.; Klooster, J.; van Rossum, A.G.; van de Pavert, S.A.; Versteeg, I.; Cardozo, B.N.; Tonagel, F.; Beck, S.C.; Tanimoto, N.; et al. Mpp4 recruits Psd95 and Veli3 towards the photoreceptor synapse. Hum. Mol. Genet. 2006, 15, 1291–1302. [Google Scholar] [CrossRef]
  71. Bohl, J.; Brimer, N.; Lyons, C.; Vande Pol, S.B. The stardust family protein MPP7 forms a tripartite complex with LIN7 and DLG1 that regulates the stability and localization of DLG1 to cell junctions. J. Biol. Chem. 2007, 282, 9392–9400. [Google Scholar] [CrossRef] [PubMed]
  72. Stohr, H.; Molday, L.L.; Molday, R.S.; Weber, B.H.; Biedermann, B.; Reichenbach, A.; Kramer, F. Membrane-associated guanylate kinase proteins MPP4 and MPP5 associate with Veli3 at distinct intercellular junctions of the neurosensory retina. J. Comp. Neurol. 2005, 481, 31–41. [Google Scholar] [CrossRef] [PubMed]
  73. Feng, W.; Long, J.F.; Fan, J.S.; Suetake, T.; Zhang, M. The tetrameric L27 domain complex as an organization platform for supramolecular assemblies. Nat. Struct. Mol. Biol. 2004, 11, 475–480. [Google Scholar] [CrossRef]
  74. Harris, B.Z.; Venkatasubrahmanyam, S.; Lim, W.A. Coordinated folding and association of the LIN-2, -7 (L27) domain. An obligate heterodimerization involved in assembly of signaling and cell polarity complexes. J. Biol. Chem. 2002, 277, 34902–34908. [Google Scholar] [CrossRef]
  75. Petrosky, K.Y.; Ou, H.D.; Lohr, F.; Dotsch, V.; Lim, W.A. A general model for preferential hetero-oligomerization of LIN-2/7 domains: Mechanism underlying directed assembly of supramolecular signaling complexes. J. Biol. Chem. 2005, 280, 38528–38536. [Google Scholar] [CrossRef] [PubMed]
  76. Kamberov, E.; Makarova, O.; Roh, M.; Liu, A.; Karnak, D.; Straight, S.; Margolis, B. Molecular cloning and characterization of Pals, proteins associated with mLin-7. J. Biol. Chem. 2000, 275, 11425–11431. [Google Scholar] [CrossRef]
  77. Ozcelik, M.; Cotter, L.; Jacob, C.; Pereira, J.A.; Relvas, J.B.; Suter, U.; Tricaud, N. Pals1 is a major regulator of the epithelial-like polarization and the extension of the myelin sheath in peripheral nerves. J. Neurosci. 2010, 30, 4120–4131. [Google Scholar] [CrossRef]
  78. Zollinger, D.R.; Chang, K.J.; Baalman, K.; Kim, S.; Rasband, M.N. The polarity protein Pals1 regulates radial sorting of axons. J. Neurosci. 2015, 35, 10474–10484. [Google Scholar] [CrossRef]
  79. Yamada, T.; Saitoh, Y.; Kametani, K.; Kamijo, A.; Sakamoto, T.; Terada, N. Involvement of membrane palmitoylated protein 2 (MPP2) in the synaptic molecular complex at the mouse cerebellar glomerulus. Histochem. Cell Biol. 2022, 158, 497–511. [Google Scholar] [CrossRef] [PubMed]
  80. Kamijo, A.; Saitoh, Y.; Sakamoto, T.; Kubota, H.; Yamauchi, J.; Terada, N. Scaffold protein Lin7 family in membrane skeletal protein complex in mouse seminiferous tubules. Histochem. Cell Biol. 2019, 152, 333–343. [Google Scholar] [CrossRef] [PubMed]
  81. Monzani, E.; Bazzotti, R.; Perego, C.; La Porta, C.A. AQP1 is not only a water channel: It contributes to cell migration through Lin7/β-catenin. PLoS ONE 2009, 4, e6167. [Google Scholar] [CrossRef]
  82. Perego, C.; Vanoni, C.; Massari, S.; Longhi, R.; Pietrini, G. Mammalian LIN-7 PDZ proteins associate with β-catenin at the cell-cell junctions of epithelia and neurons. EMBO J. 2000, 19, 3978–3989. [Google Scholar] [CrossRef] [PubMed]
  83. Ito, H.; Morishita, R.; Nagata, K.I. Functions of Rhotekin, an effector of Rho GTPase, and its binding partners in mammals. Int. J. Mol. Sci. 2018, 19, 2121. [Google Scholar] [CrossRef] [PubMed]
  84. Massari, S.; Perego, C.; Padovano, V.; D’Amico, A.; Raimondi, A.; Francolini, M.; Pietrini, G. LIN7 mediates the recruitment of IRSp53 to tight junctions. Traffic 2009, 10, 246–257. [Google Scholar] [CrossRef] [PubMed]
  85. Burette, A.C.; Park, H.; Weinberg, R.J. Postsynaptic distribution of IRSp53 in spiny excitatory and inhibitory neurons. J. Comp. Neurol. 2014, 522, 2164–2178. [Google Scholar] [CrossRef]
  86. Sawallisch, C.; Berhorster, K.; Disanza, A.; Mantoani, S.; Kintscher, M.; Stoenica, L.; Dityatev, A.; Sieber, S.; Kindler, S.; Morellini, F.; et al. The insulin receptor substrate of 53 kDa (IRSp53) limits hippocampal synaptic plasticity. J. Biol. Chem. 2009, 284, 9225–9236. [Google Scholar] [CrossRef]
  87. Setou, M.; Nakagawa, T.; Seog, D.H.; Hirokawa, N. Kinesin superfamily motor protein KIF17 and mLin-10 in NMDA receptor-containing vesicle transport. Science 2000, 288, 1796–1802. [Google Scholar] [CrossRef] [PubMed]
  88. Jo, K.; Derin, R.; Li, M.; Bredt, D.S. Characterization of MALS/Velis-1, -2, and -3: A family of mammalian LIN-7 homologs enriched at brain synapses in association with the postsynaptic density-95/NMDA receptor postsynaptic complex. J. Neurosci. 1999, 19, 4189–4199. [Google Scholar] [CrossRef]
  89. Gauthier, K.D.; Rocheleau, C.E. LIN-10 can promote LET-23 EGFR signaling and trafficking independently of LIN-2 and LIN-7. Mol. Biol. Cell 2021, 32, 788–799. [Google Scholar] [CrossRef]
  90. Andersen, D.S.; Colombani, J.; Palmerini, V.; Chakrabandhu, K.; Boone, E.; Rothlisberger, M.; Toggweiler, J.; Basler, K.; Mapelli, M.; Hueber, A.O.; et al. The Drosophila TNF receptor Grindelwald couples loss of cell polarity and neoplastic growth. Nature 2015, 522, 482–486. [Google Scholar] [CrossRef]
  91. Olsen, O.; Wade, J.B.; Morin, N.; Bredt, D.S.; Welling, P.A. Differential localization of mammalian Lin-7 (MALS/Veli) PDZ proteins in the kidney. Am. J. Physiol. Renal Physiol. 2005, 288, F345–F352. [Google Scholar] [CrossRef] [PubMed]
  92. Misawa, H.; Kawasaki, Y.; Mellor, J.; Sweeney, N.; Jo, K.; Nicoll, R.A.; Bredt, D.S. Contrasting localizations of MALS/LIN-7 PDZ proteins in brain and molecular compensation in knockout mice. J. Biol. Chem. 2001, 276, 9264–9272. [Google Scholar] [CrossRef]
  93. Matsumoto, A.; Mizuno, M.; Hamada, N.; Nozaki, Y.; Jimbo, E.F.; Momoi, M.Y.; Nagata, K.; Yamagata, T. LIN7A depletion disrupts cerebral cortex development, contributing to intellectual disability in 12q21-deletion syndrome. PLoS ONE 2014, 9, e92695. [Google Scholar] [CrossRef] [PubMed]
  94. Mizuno, M.; Matsumoto, A.; Hamada, N.; Ito, H.; Miyauchi, A.; Jimbo, E.F.; Momoi, M.Y.; Tabata, H.; Yamagata, T.; Nagata, K. Role of an adaptor protein Lin-7B in brain development: Possible involvement in autism spectrum disorders. J. Neurochem. 2015, 132, 61–69. [Google Scholar] [CrossRef] [PubMed]
  95. Hara, T.; Saeki, K.; Jinnouchi, H.; Kazuno, S.; Miura, Y.; Yokomizo, T. The C-terminal region of BLT2 restricts its localization to the lateral membrane in a LIN7C-dependent manner. FASEB J. 2021, 35, e21364. [Google Scholar] [CrossRef]
  96. Bachmann, A.; Grawe, F.; Johnson, K.; Knust, E. Drosophila Lin-7 is a component of the Crumbs complex in epithelia and photoreceptor cells and prevents light-induced retinal degeneration. Eur. J. Cell Biol. 2008, 87, 123–136. [Google Scholar] [CrossRef]
  97. Hori, K.; Konno, D.; Maruoka, H.; Sobue, K. MALS is a binding partner of IRSp53 at cell-cell contacts. FEBS Lett. 2003, 554, 30–34. [Google Scholar] [CrossRef]
  98. Simske, J.S.; Kaech, S.M.; Harp, S.A.; Kim, S.K. LET-23 receptor localization by the cell junction protein LIN-7 during C. elegans vulval induction. Cell 1996, 85, 195–204. [Google Scholar] [CrossRef]
  99. Ide, N.; Hirao, K.; Hata, Y.; Takeuchi, M.; Irie, M.; Yao, I.; Deguchi, M.; Toyoda, A.; Nishioka, H.; Mizoguchi, A.; et al. Molecular cloning and characterization of rat lin-10. Biochem. Biophys. Res. Commun. 1998, 243, 634–638. [Google Scholar] [CrossRef]
  100. Tseng, T.C.; Marfatia, S.M.; Bryant, P.J.; Pack, S.; Zhuang, Z.; O’Brien, J.E.; Lin, L.; Hanada, T.; Chishti, A.H. VAM-1: A new member of the MAGUK family binds to human Veli-1 through a conserved domain. Biochim. Biophys. Acta 2001, 1518, 249–259. [Google Scholar] [CrossRef]
  101. Sudo, K.; Ito, H.; Iwamoto, I.; Morishita, R.; Asano, T.; Nagata, K. Identification of a cell polarity-related protein, Lin-7B, as a binding partner for a Rho effector, Rhotekin, and their possible interaction in neurons. Neurosci. Res. 2006, 56, 347–355. [Google Scholar] [CrossRef] [PubMed]
  102. Shingai, T.; Ikeda, W.; Kakunaga, S.; Morimoto, K.; Takekuni, K.; Itoh, S.; Satoh, K.; Takeuchi, M.; Imai, T.; Monden, M.; et al. Implications of nectin-like molecule-2/IGSF4/RA175/SgIGSF/TSLC1/SynCAM1 in cell-cell adhesion and transmembrane protein localization in epithelial cells. J. Biol. Chem. 2003, 278, 35421–35427. [Google Scholar] [CrossRef] [PubMed]
  103. Biederer, T.; Sara, Y.; Mozhayeva, M.; Atasoy, D.; Liu, X.; Kavalali, E.T.; Sudhof, T.C. SynCAM, a synaptic adhesion molecule that drives synapse assembly. Science 2002, 297, 1525–1531. [Google Scholar] [CrossRef]
  104. Fukuhara, H.; Masuda, M.; Yageta, M.; Fukami, T.; Kuramochi, M.; Maruyama, T.; Kitamura, T.; Murakami, Y. Association of a lung tumor suppressor TSLC1 with MPP3, a human homologue of Drosophila tumor suppressor Dlg. Oncogene 2003, 22, 6160–6165. [Google Scholar] [CrossRef] [PubMed]
  105. Rademacher, N.; Schmerl, B.; Lardong, J.A.; Wahl, M.C.; Shoichet, S.A. MPP2 is a postsynaptic MAGUK scaffold protein that links SynCAM1 cell adhesion molecules to core components of the postsynaptic density. Sci. Rep. 2016, 6, 35283. [Google Scholar] [CrossRef] [PubMed]
  106. Fujita, E.; Tanabe, Y.; Imhof, B.A.; Momoi, M.Y.; Momoi, T. A complex of synaptic adhesion molecule CADM1, a molecule related to autism spectrum disorder, with MUPP1 in the cerebellum. J. Neurochem. 2012, 123, 886–894. [Google Scholar] [CrossRef] [PubMed]
  107. Jakab, R.L.; Hamori, J. Quantitative morphology and synaptology of cerebellar glomeruli in the rat. Anat. Embryol. 1988, 179, 81–88. [Google Scholar] [CrossRef]
  108. Fukaya, M.; Watanabe, M. Improved immunohistochemical detection of postsynaptically located PSD-95/SAP90 protein family by protease section pretreatment: A study in the adult mouse brain. J. Comp. Neurol. 2000, 426, 572–586. [Google Scholar] [CrossRef]
  109. Bissen, D.; Foss, F.; Acker-Palmer, A. AMPA receptors and their minions: Auxiliary proteins in AMPA receptor trafficking. Cell Mol. Life Sci. 2019, 76, 2133–2169. [Google Scholar] [CrossRef]
  110. Won, S.; Levy, J.M.; Nicoll, R.A.; Roche, K.W. MAGUKs: Multifaceted synaptic organizers. Curr. Opin. Neurobiol. 2017, 43, 94–101. [Google Scholar] [CrossRef]
  111. Yamada, K.; Fukaya, M.; Shimizu, H.; Sakimura, K.; Watanabe, M. NMDA receptor subunits GluRε1, GluRε3 and GluRζ1 are enriched at the mossy fibre-granule cell synapse in the adult mouse cerebellum. Eur. J. Neurosci. 2001, 13, 2025–2036. [Google Scholar] [CrossRef] [PubMed]
  112. Petralia, R.S.; Wang, Y.X.; Wenthold, R.J. NMDA receptors and PSD-95 are found in attachment plaques in cerebellar granular layer glomeruli. Eur. J. Neurosci. 2002, 15, 583–587. [Google Scholar] [CrossRef]
  113. Yamazaki, M.; Fukaya, M.; Hashimoto, K.; Yamasaki, M.; Tsujita, M.; Itakura, M.; Abe, M.; Natsume, R.; Takahashi, M.; Kano, M.; et al. TARPs γ-2 and γ-7 are essential for AMPA receptor expression in the cerebellum. Eur. J. Neurosci. 2010, 31, 2204–2220. [Google Scholar] [CrossRef]
  114. Kita, K.; Albergaria, C.; Machado, A.S.; Carey, M.R.; Muller, M.; Delvendahl, I. GluA4 facilitates cerebellar expansion coding and enables associative memory formation. Elife 2021, 10, e65152. [Google Scholar] [CrossRef] [PubMed]
  115. Schmerl, B.; Gimber, N.; Kuropka, B.; Stumpf, A.; Rentsch, J.; Kunde, S.A.; von Sivers, J.; Ewers, H.; Schmitz, D.; Freund, C.; et al. The synaptic scaffold protein MPP2 interacts with GABAA receptors at the periphery of the postsynaptic density of glutamatergic synapses. PLoS Biol. 2022, 20, e3001503. [Google Scholar] [CrossRef] [PubMed]
  116. Sieghart, W.; Chiou, L.C.; Ernst, M.; Fabjan, J.; Savić, M.M.; Lee, M.T. α6-Containing GABAA Receptors: Functional roles and therapeutic potentials. Pharmacol. Rev. 2022, 74, 238–270. [Google Scholar] [CrossRef]
  117. Tomita, S. Molecular constituents and localization of the ionotropic GABA receptor complex in vivo. Curr. Opin. Neurobiol. 2019, 57, 81–86. [Google Scholar] [CrossRef]
  118. Shin, S.M.; Skaar, S.; Danielson, E.; Lee, S.H. Aberrant expression of S-SCAM causes the loss of GABAergic synapses in hippocampal neurons. Sci. Rep. 2020, 10, 83. [Google Scholar] [CrossRef]
  119. Chen, M.; Koopmans, F.; Paliukhovich, I.; van der Spek, S.J.F.; Dong, J.; Smit, A.B.; Li, K.W. Blue native PAGE-antibody shift in conjunction with mass spectrometry to reveal protein subcomplexes: Detection of a cerebellar α1/α6-Subunits containing γ-aminobutyric acid type A receptor subtype. Int. J. Mol. Sci. 2023, 24, 7632. [Google Scholar] [CrossRef]
  120. Peng, Z.; Zhang, N.; Chandra, D.; Homanics, G.E.; Olsen, R.W.; Houser, C.R. Altered localization of the δ subunit of the GABAA receptor in the thalamus of α4 subunit knockout mice. Neurochem. Res. 2014, 39, 1104–1117. [Google Scholar] [CrossRef]
  121. Kralic, J.E.; Sidler, C.; Parpan, F.; Homanics, G.E.; Morrow, A.L.; Fritschy, J.M. Compensatory alteration of inhibitory synaptic circuits in cerebellum and thalamus of γ-aminobutyric acid type A receptor α1 subunit knockout mice. J. Comp. Neurol. 2006, 495, 408–421. [Google Scholar] [CrossRef] [PubMed]
  122. Tseng, W.C.; Jenkins, P.M.; Tanaka, M.; Mooney, R.; Bennett, V. Giant ankyrin-G stabilizes somatodendritic GABAergic synapses through opposing endocytosis of GABAA receptors. Proc. Natl. Acad. Sci. USA 2015, 112, 1214–1219. [Google Scholar] [CrossRef] [PubMed]
  123. Loebrich, S.; Bahring, R.; Katsuno, T.; Tsukita, S.; Kneussel, M. Activated radixin is essential for GABAA receptor α5 subunit anchoring at the actin cytoskeleton. EMBO J. 2006, 25, 987–999. [Google Scholar] [CrossRef]
  124. Zhou, X.; Gueydan, M.; Jospin, M.; Ji, T.; Valfort, A.; Pinan-Lucarre, B.; Bessereau, J.L. The netrin receptor UNC-40/DCC assembles a postsynaptic scaffold and sets the synaptic content of GABAA receptors. Nat. Commun. 2020, 11, 2674. [Google Scholar] [CrossRef]
  125. Lin, E.I.; Jeyifous, O.; Green, W.N. CASK regulates SAP97 conformation and its interactions with AMPA and NMDA receptors. J. Neurosci. 2013, 33, 12067–12076. [Google Scholar] [CrossRef]
  126. Ye, F.; Zeng, M.; Zhang, M. Mechanisms of MAGUK-mediated cellular junctional complex organization. Curr. Opin. Struct. Biol. 2018, 48, 6–15. [Google Scholar] [CrossRef]
  127. Coley, A.A.; Gao, W.J. PSD95: A synaptic protein implicated in schizophrenia or autism? Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 82, 187–194. [Google Scholar] [CrossRef] [PubMed]
  128. Frank, R.A.; Grant, S.G. Supramolecular organization of NMDA receptors and the postsynaptic density. Curr. Opin. Neurobiol. 2017, 45, 139–147. [Google Scholar] [CrossRef] [PubMed]
  129. Fujita-Jimbo, E.; Tanabe, Y.; Yu, Z.; Kojima, K.; Mori, M.; Li, H.; Iwamoto, S.; Yamagata, T.; Momoi, M.Y.; Momoi, T. The association of GPR85 with PSD-95-neuroligin complex and autism spectrum disorder: A molecular analysis. Mol. Autism 2015, 6, 17. [Google Scholar] [CrossRef] [PubMed]
  130. Ganapathiraju, M.K.; Thahir, M.; Handen, A.; Sarkar, S.N.; Sweet, R.A.; Nimgaonkar, V.L.; Loscher, C.E.; Bauer, E.M.; Chaparala, S. Schizophrenia interactome with 504 novel protein-protein interactions. NPJ Schizophr. 2016, 2, 16012. [Google Scholar] [CrossRef]
  131. Mullins, N.; Forstner, A.J.; O’Connell, K.S.; Coombes, B.; Coleman, J.R.I.; Qiao, Z.; Als, T.D.; Bigdeli, T.B.; Borte, S.; Bryois, J.; et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat. Genet. 2021, 53, 817–829. [Google Scholar] [CrossRef] [PubMed]
  132. Cross-Disorder Group of the Psychiatric Genomics Consortium. Genomic relationships, novel loci, and pleiotropic mechanisms across eight psychiatric disorders. Cell 2019, 179, 1469–1482.e11. [Google Scholar] [CrossRef] [PubMed]
  133. Schizophrenia Working Group of the Psychiatric Genomics Consortium. Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014, 511, 421–427. [Google Scholar] [CrossRef] [PubMed]
  134. Tobler, R.; Souilmi, Y.; Huber, C.D.; Bean, N.; Turney, C.S.M.; Grey, S.T.; Cooper, A. The role of genetic selection and climatic factors in the dispersal of anatomically modern humans out of Africa. Proc. Natl. Acad. Sci. USA 2023, 120, e2213061120. [Google Scholar] [CrossRef]
  135. Khoury, S.; Wang, Q.P.; Parisien, M.; Gris, P.; Bortsov, A.V.; Linnstaedt, S.D.; McLean, S.A.; Tungate, A.S.; Sofer, T.; Lee, J.; et al. Multi-ethnic GWAS and meta-analysis of sleep quality identify MPP6 as a novel gene that functions in sleep center neurons. Sleep 2021, 44, zsaa211. [Google Scholar] [CrossRef] [PubMed]
  136. Grant, S.G. The molecular evolution of the vertebrate behavioural repertoire. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2016, 371, 20150051. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of an erythrocyte membrane skeleton. The spectrin–actin network structure is connected by protein 4.1R-membrane palmitoylated protein 1 (MPP1) and ankyrin to the intramembranous proteins glycophorin C (GPC) and band 3, respectively. The concept was obtained from previous research [4].
Figure 1. Schematic representation of an erythrocyte membrane skeleton. The spectrin–actin network structure is connected by protein 4.1R-membrane palmitoylated protein 1 (MPP1) and ankyrin to the intramembranous proteins glycophorin C (GPC) and band 3, respectively. The concept was obtained from previous research [4].
Genes 14 01942 g001
Figure 2. Schematic representation of the relationships among membrane skeletal proteins (4.1, MPP, and CADM) in the PNS (a), CNS (b), and testis (c). Note the different interdependences among those proteins in different organs, revealed by the genetic depletion of the proteins. The picture is partially modified from a previous paper [51].
Figure 2. Schematic representation of the relationships among membrane skeletal proteins (4.1, MPP, and CADM) in the PNS (a), CNS (b), and testis (c). Note the different interdependences among those proteins in different organs, revealed by the genetic depletion of the proteins. The picture is partially modified from a previous paper [51].
Genes 14 01942 g002
Figure 3. (a): Schematic representation of MPP2-relating proteins in the cerebellar glomerulus. Note that MPP2 interacts with various adhesion molecules, such as CADM1 and M-cadherin, as well as signal transduction proteins such as CASK and Lin7. GAD: glutamic acid decarboxylase, VGLUT1: vesicular glutamate transporter 1. The picture is partially modified from a previous paper [79]. (bk): Localization of MAGUKs (MPP2 (a,f), DLG2 (b,g), PSD95 (c,h), CASK (d,i), and SAP97 (Dlg1) (e,j)) in the cerebellar cortex in MPP2+/+ (ae) and MPP-/- (fj) mice. Note that MPP2 is mainly observed in the granular layer (GL). ML: molecular layer, PCL: Purkinje cell layer.
Figure 3. (a): Schematic representation of MPP2-relating proteins in the cerebellar glomerulus. Note that MPP2 interacts with various adhesion molecules, such as CADM1 and M-cadherin, as well as signal transduction proteins such as CASK and Lin7. GAD: glutamic acid decarboxylase, VGLUT1: vesicular glutamate transporter 1. The picture is partially modified from a previous paper [79]. (bk): Localization of MAGUKs (MPP2 (a,f), DLG2 (b,g), PSD95 (c,h), CASK (d,i), and SAP97 (Dlg1) (e,j)) in the cerebellar cortex in MPP2+/+ (ae) and MPP-/- (fj) mice. Note that MPP2 is mainly observed in the granular layer (GL). ML: molecular layer, PCL: Purkinje cell layer.
Genes 14 01942 g003
Figure 4. Comparative localization of GABAARα1 (a,f,k) with MPP2 (b,c), gephyrin (g,h) and GABAARα6 (l,m) in mouse cerebellar glomeruli. Examples of two-color overlapping regions are shown in (d,e,i,j,n,o) from areas in pictures (c,h,m), respectively. Detailed count data regarding the overlap is described in the text. The right lane demonstrates a summarized schematic drawing of their localizations obtained by immunohistochemistry; it does not consider how to make GABAAR with five subunits.
Figure 4. Comparative localization of GABAARα1 (a,f,k) with MPP2 (b,c), gephyrin (g,h) and GABAARα6 (l,m) in mouse cerebellar glomeruli. Examples of two-color overlapping regions are shown in (d,e,i,j,n,o) from areas in pictures (c,h,m), respectively. Detailed count data regarding the overlap is described in the text. The right lane demonstrates a summarized schematic drawing of their localizations obtained by immunohistochemistry; it does not consider how to make GABAAR with five subunits.
Genes 14 01942 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Terada, N.; Saitoh, Y.; Saito, M.; Yamada, T.; Kamijo, A.; Yoshizawa, T.; Sakamoto, T. Recent Progress on Genetically Modified Animal Models for Membrane Skeletal Proteins: The 4.1 and MPP Families. Genes 2023, 14, 1942. https://doi.org/10.3390/genes14101942

AMA Style

Terada N, Saitoh Y, Saito M, Yamada T, Kamijo A, Yoshizawa T, Sakamoto T. Recent Progress on Genetically Modified Animal Models for Membrane Skeletal Proteins: The 4.1 and MPP Families. Genes. 2023; 14(10):1942. https://doi.org/10.3390/genes14101942

Chicago/Turabian Style

Terada, Nobuo, Yurika Saitoh, Masaki Saito, Tomoki Yamada, Akio Kamijo, Takahiro Yoshizawa, and Takeharu Sakamoto. 2023. "Recent Progress on Genetically Modified Animal Models for Membrane Skeletal Proteins: The 4.1 and MPP Families" Genes 14, no. 10: 1942. https://doi.org/10.3390/genes14101942

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop