Next Article in Journal
Nanoplatform for the Delivery of Topotecan in the Cancer Milieu: An Appraisal of its Therapeutic Efficacy
Next Article in Special Issue
Bone Metabolism Effects of Medical Therapy in Advanced Renal Cell Carcinoma
Previous Article in Journal
RUNAT-BI: A Ruthenium(III) Complex as a Selective Anti-Tumor Drug Candidate against Highly Aggressive Cancer Cell Lines
Previous Article in Special Issue
Regulation of Chromatin Accessibility by the Farnesoid X Receptor Is Essential for Circadian and Bile Acid Homeostasis In Vivo
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endosomal Sorting Protein SNX27 and Its Emerging Roles in Human Cancers

1
Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
2
Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA
3
University of Illinois at Chicago (UIC) Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
4
Jesse Brown VA Medical Center, Chicago, IL 60612, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(1), 70; https://doi.org/10.3390/cancers15010070
Submission received: 10 November 2022 / Revised: 14 December 2022 / Accepted: 18 December 2022 / Published: 22 December 2022
(This article belongs to the Special Issue At the Interface of Genetics, Epigenetics and Metabolism in Cancer)

Abstract

:

Simple Summary

The sorting nexin (SNX) family of proteins mediate the sorting and trafficking of endocytosed transmembrane proteins between the endosomal compartments, lysosome, trans-Golgi network, and plasma membrane. SNXs are characterized by the presence of a Phox domain; however, SNX27 is a unique member of this family as it contains an additional PDZ domain. Evidence suggests that SNX27, in association with the retromer complex, binds cargo via its PDZ domain and recycles them from the early endosomes to the plasma membrane, thereby escaping their lysosomal degradation. The roles of SNX27 in cancer is largely unexplored, however many of its identified cargoes have been indicated in tumorigenesis. In this review we provide a summary of the current knowledge, as supported by scientific literature and evidence, regarding the fundamental structure, biological function, and implication of SNX27 in human cancers.

Abstract

SNX27 belongs to the sorting nexin (SNX) family of proteins that play a critical role in protein sorting and trafficking in the endocytosis pathway. This protein family is characterized by the presence of a Phox (PX) domain; however, SNX27 is unique in containing an additional PDZ domain. Recently, SNX27 has gained popularity as an important sorting protein that is associated with the retromer complex and mediates the recycling of internalized proteins from endosomes to the plasma membrane in a PDZ domain-dependent manner. Over 100 cell surface proteins have been identified as binding partners of the SNX27–retromer complex. However, the roles and underlying mechanisms governed by SNX27 in tumorigenesis remains to be poorly understood. Many of its known binding partners include several G-protein coupled receptors, such as β2-andrenergic receptor and parathyroid hormone receptor, are associated with multiple pathways implicated in oncogenic signaling and tumorigenesis. Additionally, SNX27 mediates the recycling of GLUT1 and the activation of mTORC1, both of which can regulate intracellular energy balance and promote cell survival and proliferation under conditions of nutrient deprivation. In this review, we summarize the structure and fundamental roles of SNX proteins, with a focus on SNX27, and provide the current evidence indicating towards the role of SNX27 in human cancers. We also discuss the gap in the field and future direction of SNX27 research. Insights into the emerging roles and mechanism of SNX27 in cancers will provide better development strategies to prevent and treat tumorigenesis.

Graphical Abstract

1. Introduction

SNX27 belongs to the large family of SNX proteins whose fundamental role involves the regulation of intracellular trafficking, as well as sorting of internalized cargo through the complicated cascade of the endocytic pathway [1]. The SNX proteins are mainly characterized by the presence of a specific phospholipid binding Phox (PX) homology domain [2]. However, the presence or absence of additional domains further categorize the SNX proteins into different subgroups as they regulate distinct functions in target binding and specificity [3]. This includes the SNX-BAR subfamily that consists of an additional BAR (Bin/Amphiphysin/Rvs) domain [4] and the SNX-FERM subfamily with additional FERM (4.1/ezrin/radixin/moesin) domain [1]. SNX27 belongs to the SNX-FERM subfamily. However, structural analysis has revealed that in addition to the PX and FERM domains, SNX27 carries a third PDZ (post-synaptic density 95/discs large/zonula occludens-1) binding domain. This makes SNX27 the most unique member of the SNX family because it mediates the rescue of endocytosed transmembrane proteins from lysosomal degradation in a PDZ-domain dependent manner [5].
Endocytosis is a crucial cellular process that internalizes cargoes from the cell surface and transports them in membrane-bound vesicles for sorting into either of its two destined fates: lysosomal degradation or recycling back to the plasma membrane [6]. Improper sorting of internalized cargo and defects in the endosomal recycling pathway have been linked to a variety of human diseases, such as neurodegeneration disorders as well as cancer [7]. However, the underlying mechanisms governing endosomal cargo sorting, retrieval and recycling, and its implication in disease pathophysiology is still poorly understood.
Retromer is an endosomal coat protein complex first identified in the endolysosomal system of budding yeast [8]. It is evolutionarily conserved in mammals and has been implicated in the sorting and transport of several transmembrane proteins from early endosomes to either the trans-Golgi network (TGN) or the cell surface, thus rescuing the cargo from lysosomal degradation [9]. The mammalian retromer complex consists of a constitutive heterotrimer of VPS26-VPS29-VPS35 proteins that has been shown to assist in cargo recognition [10]. Several members of the SNX family of proteins have been identified to associate with the retromer complex, such as the SNX1/2 and SNX5/6 heterodimer of the SNX-BAR subfamily [4]; the monomeric SNX3 of the SNX-PX subfamily [11]; and the most recently identified associate, SNX27 [1]. While the SNX-BAR heterodimers assist in retromer-mediated endosomal membrane remodeling [10], SNX3 and SNX27 have been suggested to act as cargo adaptors [1]. However, the association of SNX27 and the retromer complex is critical for the cell surface recycling of transmembrane proteins carrying a PDZ-binding motif [3]. Therefore, the emerging roles of the SNX27-retromer complex has gained recognition in the field of recycling of major solute carriers, cell surface receptors, and other transmembrane proteins. However, the mechanisms driving SNX27 mediated endosomal recycling of proteins remains largely unknown.
Interactions between the intracellular compartments and the extracellular environment need to be closely monitored in order to maintain cellular homeostasis [12]. Therefore, improper sorting of endocytosed cargo can disrupt the balance between the cell surface and intracellular expression levels of transmembrane proteins, which may contribute to disease progression and pathophysiology, if left unchecked.
Emerging evidence suggests a potential role of SNXs in human cancers, although the research is still limited. This review is aimed towards summarizing scientific evidence supporting and providing an update on the current molecular understanding of SNX27, its associations with the sorting and transport of internalized endosomal cargo, and its implication in cancers.

2. Overview of the Sorting Nexin Family of Proteins

Since the discovery of the first sorting nexin, a total of 10 family members have been identified in yeast and 33 members in mammals [13]. Typically, the sequence length of SNX proteins range between 400–700aa with a common PX domain, which is approximately 100–130aa long [14]. Since PX domains have high binding affinities to phosphatidylinositol 3-monophosphates (PtdIns3P), SNX proteins are attracted to organelle membranes rich in these phospholipids, such as the endosomal membrane [15]. However, some members of the SNX family have additional structural domains that have shown to regulate their target specificity, intracellular localization, as well as membrane conformation, which also gives rise to various SNX subfamilies (see Table 1) [2].
One of the most critical physiological roles of SNX proteins that has been studied so far is their ability to mediate the trafficking of endocytosed cargoes from the compartments of early endosomes to either the TGN or the plasma membrane [16]. However, it has been established that the successful recycling and sorting of endocytosed cargoes is dependent on the association of SNX proteins with the retromer complex, a heterotrimer of VPS35, VPS26 and VPS29 subunits [17]. In the mammalian system, three different SNX-retromer associations have been identified which include the SNX3-, SNX-BAR-, and SNX27-retromer complexes [18].
SNX3 belongs to the SNX-PX subfamily of SNX proteins as it contains only the PX domain. The association of SNX3 with the retromer subunit, VPS26, has been shown to be crucial for cargo recognition, binding, and retrograde recycling to the TGN [10]. This is due to the fact that the SNX3/VPS26 interaction forms a binding site at its interface for cargoes enriched in L-M-V domain, while the SNX3-PX domain exclusively interacts with the cytosolic face of early endosomes [19]. Some of the SNX3-retromer cargoes that have been identified include Wntless, a Wnt sorting receptor [20], and Dmt-II metal ion transporter [21].
In mammals, up to twelve members of the SNX-BAR subfamily have been identified which possess a Bin/Amphiphysin/Rvs (BAR) domain in addition to the highly conserved PX domain [22]. Among these twelve members, there are high structural similarities between SNX1 and 2, as well as SNX5 and 6, which form a functional SNX1/2:SNX5/6 heterodimer, representing their yeast counterpart Vps5:Vps17 [13]. Further analysis revealed that SNX1, the first SNX protein to be identified in mammals, binds to the retromer and mediates the formation of the SNX-BAR-retromer complex consisting of the SNX1/2: SNX5/6 heterodimer subunit [23]. This association further promotes the membrane remodeling and tubule formation necessary for promoting the retrograde transport of selected cargoes, such as the mannose-6-phosphate receptor, to the TGN [24].
SNX27 was identified as another retromer associated SNX protein belonging to the SNX-FERM subfamily. Members of this subfamily contain a PX domain and a C-terminal 4.1/ezrin/radixin/moesin (FERM) domain [25]. The mechanisms of SNX27-mediated endocytic recycling of proteins has not been fully elucidated yet; however, the current understanding suggests that the SNX27-retromer complex assists in the direct trafficking of cargo from the endosomal compartments to the plasma membrane, and that it requires an association with the SNX-BAR subunit [26]. Furthermore, SNX27 is a unique member of the SNX family as it is the only known SNX protein carrying an additional N-terminal post-synaptic density 95/discs large/zonula occludens-1 (PDZ) domain, and current models suggest that the SNX27-retromer complex recognizes and binds to target proteins in a PDZ-domain dependent manner [3].
In addition to their primary role in endocytic recycling, some SNX proteins are involved in the process of endocytosis and lysosomal degradation. SNX9 and SNX18 carry an N-terminal SH3 domain in addition to the C-terminal BAR domain [27]. While SNX9 can directly bind to dynamin-1/2 and regulate clathrin-mediated endocytosis, SNX18 has been particularly shown to compensate for the loss of SNX9 during endocytosis of transferrin receptors [28]. Meanwhile, SNX1, SNX6 and SNX11 have shown to assist in the sorting and lysosomal degradation of epidermal growth factor receptor (EGFR) [23], tumor suppressor p27Kip1 [29], and TRPV3 [30], respectively.

3. Structure and Function of SNX27

3.1. SNX27 and Its Special Domains

As shown in Figure 1, SNX27 contains three distinct domains: a central PX domain, a C-terminal FERM domain, and an N-terminal PDZ domain (only in SNX27) [2] (Figure 1). Despite carrying a common PX and FERM domain, SNX27 behaves functionally distinct from the other members of the SNX-FERM subfamily [25]. While both SNX17 and SNX31 depend on their FERM domain and have strong affinity towards targeting and recycling cargoes carrying a NPxY/NxxY motif, SNX27 mediates endocytic recycling of cargoes in a PDZ-domain dependent manner [25]. The SNX27-FERM domain associates with an N-terminal DLF motif found on the SNX1/2 homodimer, which has not been reported in either SNX17 or SNX31 [25]. This was further supported by structural analysis data, which reported very low sequence identity between SNX27-FERM domain compared with that of SNX17 and SNX31 [25]. Therefore, the FERM domain has a distinct role in SNX27 than in the other SNX-FERM members, such that it only promotes SNX27 interaction with the SNX-BAR proteins. This structure mediates the recruitment of SNX27 to the membranes of early endosomes and initiates membrane remodeling to assist in the endocytic recycling and trafficking of PDZ-domain containing cargoes [31].
SNX27 was initially discovered in the brain tissue where it was found to be interacting with 5-HT4.a/R, a G-protein coupled receptor (GPCR) [32]. Structural analyses later revealed that 5-HT4.a/R carries a C-terminal class-I PDZ-binding motif and its association with SNX27 was found to be dependent on its interaction with the PDZ-domain [33]. Therefore, the canonical basis for the PDZ domain to recognize and bind to cargoes was predisposed to the presence of a class-I PDZ-binding motif [S/T]-x-Φ (where x represents any amino acid and Φ represents any hydrophobic amino acid) at the C-termini, often present in the cytosolic tails of transmembrane proteins [34]. However, presence of certain sequences upstream of the C-terminus PDZ-binding motif was also shown to enhance binding specificity [35]. Studies have revealed that acidic side chains are often found alongside the PDZ-binding motifs, however several SNX27 cargoes have replaced these upstream acidic side chain sequences with conserved serine/threonine phosphorylation sites which was later shown to mimic the acidic side chain residues, thus promoting binding affinity in a similar manner [35]. With the advent of proteomics and sequence quantitative analysis, over hundreds of transmembrane proteins have been identified to be interacting with SNX27-PDZ domain. These include β-2-adrenergic receptor, GLUT1 glucose transporter, and the Menkes copper transporter ATP7A [36].

3.2. SNX27 Mediated Endosomal Recycling of Proteins

Endocytosed transmembrane proteins require to go through a complex network of the endomembrane system that is subdivided into the early, late, and recycling endosome vesicles. While the cargo sorted in late endosomes proceed for lysosomal degradation, those present in the recycling endosomes further interact with the components of the secretory pathway in order to be transported to either the TGN or delivered back to the cell surface [6]. Therefore, improper sorting of endocytosed cargo can disrupt physiological homeostasis and give rise to various health problems and diseases [7].
Sorting and recycling of the transmembrane proteins from early/recycling endosomal compartments directly to the plasma membrane is heavily dependent on the association of the SNX27/SNX-BAR/retromer complex on Rab4-positive endosomes [37]. In this model, the PX domain is critical for the recruitment and binding to the endosomal membrane; the FERM domain recruits SNX-BAR heterodimers which initiates membrane remodeling and tubule formation necessary for transporting the cargo [1], and the PDZ domain binds to VPS26 retromer subunit and acts as the cargo adaptor [36]. Studies have shown that the disruption of either of these components impairs SNX27 dependent cargo binding and trafficking.
While the PX domain drives SNX27 membrane recruitment, structure-based analyses have shown that the synergistic interaction of the FERM domain with the endosomal membranes also enhances localization of the SNX27-retromer complex to the endosomes, as disrupting the FERM domain decreased the affinity between SNX27 and the endosomal compartments [25].
The actin remodeling WASH complex, which consists of the WASH1, WASHC3, WASHC4, WASHC5, and FAM21 subunits [38], was identified as another SNX27-retromer interacting partner, which synergistically binds to the SNZ27-FERM domain as well as the VPS35 subunit of retromer via FAM21 [39]. Upon its recruitment, the WASH complex has shown to initiate the formation of F-actin filaments on the endosomal membranes which promotes trafficking of cargo from endosomes to the plasma membrane directly [40]. This has been particularly observed in the SNX27 PDZ-domain-dependent cell surface recycling of GLUT1, as disrupting the binding of the WASH component to the SNX27-retromer complex directed the recycling of GLUT1 to the TGN instead of the plasma membrane [26]. Therefore, this interaction may be critical for the assembly and/or regulation of the SNX27-retromer complex.
Among the many identified PDZ-domain-containing cargoes dependent on the SNX27-retromer-mediated endosome-to-plasma membrane recycling (see Table 2), β-2-andrenergic receptor was the very first transmembrane protein [36]. Some other cargoes include Ras, a monomeric small GTPase [25]; zonula occludens 2 (ZO2), an epithelial tight junction protein [36]; and AMPA receptor [41]. More recently identified cargoes include OTULIN, a deubiquitinating enzyme, which contains a class-1 PDZ-binding motif [42]. However, via X-ray crystallography it was observed that OTULIN has a high affinity to the PDZ-VPS26 binding site in addition to the canonical PDZ domain-binding motif interaction site [42]. SNX27 interactome analysis also revealed NHE3, which depends on the PDZ-domain interaction with SNX27 for its recycling from early endosomes to the plasma membrane [43]. In addition to maintaining NHE3 surface expression levels, SNX27 was also found to be necessary for brush border stability as SNX27 deletion in intestinal epithelial cells reduced NHE3 basal activity. Similarly, DRA was found to be dependent on SNX27 for its recycling to the apical plasma membrane, as shown in CaCo2 intestinal epithelial cells [44]. These studies have revealed a novel function of SNX27 mediated endocytic recycling of transmembrane proteins in the gastrointestinal tract which may be of importance in studying disease pathogenesis.
In contrast to the canonical mechanism of class-1 PDZ-binding motifs necessary for interaction with SNX27, it was recently observed that MT1-MMP, which lacks a class-1 PDZ-binding motif on its C-terminus, is still able to bind to the SNX27-retromer complex and is recycled to the cell surface in a SNX27-retomer-dependent manner [50]. Structural analysis revealed that this interaction is mediated via the DKV motif present in the cytosolic tail of MT1-MMP, which has shown to possess the features similar to a class-III PDZ-binding motif, however further biophysical studies are deemed necessary to fully understand this association [62]. This observation provides novel insights into the possibilities of non-canonical mechanisms of SNX27 PDZ-domain interactions with its cargoes.
Overall, SNX27’s interacts with various transmembrane proteins for the maintenance of their cell surface levels and activity. It suggests that loss of SNX27 may lead to an array of cellular dysfunction, which has been reciprocated in vivo as SNX27 whole-body knockout mice have been shown to be embryonically lethal [63].

3.3. SNX27 and Activation of T-cells

SNX27 is found to be expressed in almost every cell type, based on the data from Human Atlas (https://www.proteinatlas.org/ENSG00000143376-SNX27/tissue, accessed on 21 December 2022). Primarily, SNX27 was studied in the brain–nervous system and related diseases. However, we believe its role in other organs could be equally critical.
SNX27 was found to be localized at the immune synapse (IS) in a T-cell receptor (TCR) activation dependent manner [64]. Cells of the immune system give a great example of an intricate molecular network that depends on the endocytic recycling pathway to maintain constant communication between the intercellular components and the extracellular environment. The plasma membrane of T-cell lymphocytes expresses several important receptors, including TCR, which is necessary for the recognition of antigens presented by an antigen-presenting cell (APC) [65]. The binding of the TCR and a recognized antigenic peptide triggers a rapid morphological change within the T-cells causing actin remodeling and vesicular polarized trafficking of organelles towards the T cell-APC interface, thus forming the immune synapse (IS) [66]. This event is critical for sustaining intact communication between T-cell and the APC which ensures the activation of T-cell and downstream immune responses [67]. Extensive studies revealed that T-cells maintain surface level TCR expression and other signaling components necessary for IS via endosomal recycling pathways [68,69].
Under unstimulated conditions, SNX27 is predominantly expressed on the PtdIns[1]P-rich membranes of early and recycling endosomes, which then rapidly polarizes towards the IS upon TCR activation [34]. Structural studies have revealed that this redistribution is mediated by the interactions between the PX and FERM domains of SNX27 with PtdIns[1]P and PtdIns(4,5)P2/PtdIns(3,4,5)P3-enriched membranes, respectively [70,71]. This was further supported by Tello-Lafoz et al., as they showed that disruption of the FERM domain impaired SNX27 spatial distribution during IS initiation [64].
Proteomic analysis of the SNX27 interactome in IS forming activated T-cell lymphocytes has not only confirmed the participation of retromer and WASH complexes in SNX27-mediated polarized trafficking, but also revealed several SNX27 interacting cargoes that are presented at the IS in a PDZ-dependent manner, e.g., diacylglycerol kinase-ζ (DGKζ) [34,72]. TCR activation leads to the generation and accumulation of diacylglycerols (DAGs) at the IS which facilitates the recruitment of other signaling proteins involved in IS formation and maintenance [73]. While DGKζ is a negative regulator of DAG, its association with SNX27 prevents degeneration of DAG, thereby regulating IS stability via SNX27-mediated DAG metabolism [34]. Likewise, disrupting the SNX27-DGKζ interaction in activated T-cells affected downstream signaling pathways, as indicated by increased ERK phosphorylation and NF-κB hyperactivation upon either SNX27 or DGKζ silencing [34,74,75]. However, silencing of SNX27 does not affect protein expression levels of DGKζ, which suggests that the association of DGKζ with SNX27 is only necessary for its spatial distribution and trafficking during IS formation [75].
The identification of DGKζ and its association with SNX27 provides novel insights into DAG associated T-cell activation. It also suggests the tissue specific role of SNX27 in immune system. Further investigation is required to determine the extent of SNX27-mediated regulation of IS assembly and participation of other SNX27-interacting cargoes in this process. Underlying the importance of SNX27 mediated endocytic recycling of a wide array of transmembrane proteins with distinct functions, SNX27 is necessary for proper functioning of human health [76].

3.4. SNX27 in Neurodegenerative Disorders

The PDZ binding motif is most commonly found in proteins involved in excitatory synapses, and thus located within regions of postsynaptic densities in the neurons [77]. In the brain itself, SNX27 is found to be localized primarily within dendrites and has been shown to regulate synaptic plasticity [78]. SNX27 was first identified in the brain in an experiment wherein metamphetamine induced stimulation of dopamine receptors caused an upregulation of SNX27 [79]. Additionally, in vivo studies have shown that SNX27 is also critical for postnatal growth and survival as SNX27-/- mice die shortly after birth [56]. Therefore, dysregulated SNX27 functioning has been reported in several neurological and degenerative diseases, such as Alzheimer’s disease.
Multiple studies have shown enlargement of early endosomes and multivesicular bodies in brain tissues and neurons isolated from human as well murine models of Alzheimer’s disease (AD), overall suggesting defects in endocytic trafficking and recycling pathways [80,81]. SNX27 loss-of-function proteomic analysis has also demonstrated that SNX27 mediates recycling of internalized AD-related protein APP, as deletion of SNX27 decreased cell surface expression levels of APP [60,82]. However, the authors of this study were unable to detect direct binding of SNX27 to APP, suggesting the involvement of an intermediate molecule promoting SNX27/APP interaction [3]. Previously, an intracellular sorting receptor, SorLA, was identified as an APP binding protein, and reduction in SorLA expression levels caused APP cellular redistribution [83]. Therefore, functional studies looking into the interaction between SNX27, APP and SorLA were conducted, and the results revealed that SorLA binds to APP as well as SNX27 and forms a ternary structural protein complex, thereby acting as the molecular link in SNX27 mediated trafficking and recycling of APP [60].
SNX27 has also been reported to contribute towards impairment in neuronal and learning abilities in Down’s Syndrome (DS) [77]. The overall expression levels of SNX27 are shown to be downregulated in brain tissues from human patients and murine models of DS. Interestingly, rescue studies in DS mouse model have shown that overexpression of SNX27 reverses or corrects DS-related cognitive and synaptic impaired phenotypes [77]. Further analysis revealed that expression of SNX27 is regulated by C/EBPβ, a transcription factor targeted by miR-155. It was identified that miR-155 is a micro-RNA encoded on chromosome 21; therefore, trisomy of chromosome 21 in DS drives up miR-155 which causes silencing of C/EBPβ, thereby downregulating SNX27 [77]. More recently SNX27 was shown to recycle the myelination-related protein, GRP17, which is crucial for the differentiation and maturation of oligodendrocytes [61]. Improper functioning and distribution of oligodendrocytes has been reported in human and mouse DS as it leads to abnormalities in the white matter of the brain which increases defects in cognitive and motor skills [84]. Therefore, these studies suggest an underlying mechanism of SNX27 mediated neuropathogenesis of DS through oligodendrocyte dysfunction.
Lastly, SNX27 has been shown to recycle AMPA and NMDA glutamate receptors [56,78]. Glutamate serves as the primary neurotransmitter in the brain and therefore plays a critical role in mediating cognitive function and excitatory synapses, both of which promote learning and motor abilities [85]. Consequently, heterozygous SNX27-/+ mice appear to have learning and memory disabilities associated with a reduction in cell surface expression of AMPA or NMDA receptors [63]. Therefore, dysregulated expression of SNX27 can lead to seizures and epilepsy among a plethora of other neurodegenerative diseases [77].

4. Identifying the Novel Role of SNX27 in Cancer

Increasing evidence suggests that SNX27 may mediate cancer development and/or progression as it influences distinct protein–protein interactions, membrane remodeling, and cell surface expression of several important tight junction proteins and receptors implicated in cell signaling pathways (see Table 3) [35]. Human genome database analysis reveals that SNX27 is highly expressed in several cancers, in particular invasive breast cancer, which is inversely correlated with overall patient survival [86].
We have previously demonstrated that loss of SNX27 dramatically reduces tumor growth and proliferation in breast cancer cells, which was further confirmed via mouse xenograft models [87]. We further reported a novel role of SNX27 in reducing aggressiveness and invasive capacity of the cancer cells via the modulation of EMT marker Vimentin, and cell–cell junction markers, such as Claudin-5 and E-cadherin.
Another study investigating the associations of SNX27 and breast cancer metastasis revealed that SNX27 directly interacts with MT1-MMP, a key enzyme recruited by invadopodia for degradation and remodeling of the extracellular matrix [50]. The in vitro analysis demonstrated that the association of the SNX27-retromer complex is critical for the recycling of MT1-MMP to the cell surface, as deletion of either component resulted in the accumulation of MT1-MMP in late endosomes. It is also of interest to note that, unlike the other known SNX27 binding partners, MT1-MMP lacks the PDZ-binding motif. Instead, the cytosolic tail of MT1-MMP harbors a DKV motif that closely resembles a Class-III PDZ-binding motif sequence of X[DE]Xϕ, where X represents any amino acid and ϕ represents a hydrophobic residue [88]. Therefore, this study revealed a new binding partner of SNX27 harboring a Class-III PDZ-binding motif.
Several studies have revealed protein candidates from the GPCR superfamily which interact with SNX27. These include the β2-andrenergic receptor [1], parathyroid hormone receptor (PTHR) [47], metabotropic glutamate receptor 5 (mGluR5) [48], G protein-coupled inwardly rectifying potassium channel subunits (GIRK 1 and 2) [45,46], and frizzled receptor 7 (FZD7) [49]. These GPCRs are known to be involved in tumorigenesis. For example, mGluRs have been heavily reported in neuroblastoma and gliomas [89], with mGluR5 particularly driving tumorigenesis as its inhibition has shown to facilitate hypoxia-induced cell death in gliomas [90], and GIRKs are highly expressed in over 70% of non-small cell lung cancer patients which correlates with more aggressive tumors [91]. Similarly, β2-andrenergic receptor [92] and PTHR [93] have been reported to drive tumorigenesis via EMT regulation, whereas FZDs mediates tissue homeostasis and the canonical Wnt signaling pathway known to be regulated in several cancers [94]. However, there are limited studies on the tissue-special roles of SNX27 in various cancers. It is important to identify cancer-associated proteins that may interact with the PDZ-domain in a SNX27-dependent recycling and such proteins could be targeted to impair tumorigenesis.
SNX27 is involved in cellular nutrient uptake as it directly interacts with and recycles GLUT1, facilitator of glucose transport across the plasma membrane, which promotes cell growth and survival [5]. The mTORC1 activation has also been indicated to be regulated by SNX27 [3]. Although the direct mechanisms remain to be poorly understood, these studies suggest that SNX27 associations may influence cancer cell proliferation and survival by regulating intracellular energy levels.
Table 3. SNX27 associated proteins reported in cancers.
Table 3. SNX27 associated proteins reported in cancers.
TargetType of CancerRef.
β2-arBreast, gastric, pancreatic, squamous cell carcinoma[95]
GIRK2Breast, lung, hepatocellular carcinoma[91]
PTHRBreast, colorectal, prostate cancer[96]
mGluR5Gliomas, melanoma, prostate, hepatocellular carcinoma[97]
FZD7Prostate, glioma, gastric, lung, hepatocellular carcinoma[94]
ASCT2Breast, lung, prostate cancer[98]
MRP4Leukemia, liver, lung cancer[99]
SSTR5Pituitary adenomas, prostate cancer[100]
CASPBreast, pancreatic, hepatocellular carcinoma[101]
NR2CPancreatic cancer[102]
AQP2Renal cancer[103]
β-PixBreast and colorectal cancer[104]
mTORSeveral cancers: mediates cancer cell survival under stress[105]
GLUT1Several cancers: mediates nutrition uptake in cancer cells[106]
DGKζSeveral cancers: mediates immune checkpoint in cells[107]
Taken together, SNX27 may potentially exhibit its roles and regulate different stages of tumorigenesis, as we proposed in the Graphic abstract. Observations from previous studies indicate a potential therapeutic strategy of targeting SNX27 for cancer treatment, however further research is required to fully elucidate the role of SNX27 in tumorigenesis.

5. Roles of other SNX Members in Solid Cancers

Owing to the canonical physiological roles of the SNX family of proteins in mediating the endocytic pathway and intracellular protein trafficking, several SNXs have been involved in crucial intracellular signaling cascades, such as the Notch, EGFR, and Wnt signaling pathways, to name a few [108]. These pathways are known to mediate varying degrees of tumorigenicity in different types of cancer cells. Although limited, the investigation on the roles of other SNXs in different cancers has been summarized below:
A study by Zhan et al. reported that the overall mRNA and protein expression levels of SNX1 was lower in gastric cancer patients, which also correlated with poor patient survival [109]. As in vitro overexpression and knockdown studies were conducted, they observed that exogenous expression of SNX1 reduced the proliferative and migration capacity of the cancer cells. As a result, they observed an upregulation of the tumor suppressor marker E-cadherin, and downregulation of the EMT marker Vimentin, whereas the effects were reversed upon reducing the levels of SNX1. These observations are in line with another study which suggested the dependence on SNX1 for the recycling of internalized E-cadherin [110]. Therefore, a novel role for SNX1 as a putative tumor suppressor marker in gastric cancer, possibly via EMT regulation, has been suggested.
The first report of SNX5 in human cancers was identified in thyroid cancer where the expression levels of SNX5 decreased through the progression of well-differentiated to poorly differentiated tumors [111], the latter being more aggressive [112]. However, recent investigation revealed differential expression levels of SNX5 in head and neck squamous cell carcinoma where higher expression levels of SNX5 were correlated with tumor progression, aggressiveness, and poor patient survival [113]. Using mouse xenograft models, investigators revealed that loss of SNX5 reduced tumor burden by decreasing the expression levels of the anti-apoptotic protein Mcl-1 [114]. It suggests a SNX5 regulation of apoptosis in the progression of head and neck squamous cell carcinoma.
SNX6 has been reported in pancreatic cancer and implicated in regulating EMT and invasiveness of cancer cells [115]. Loss of SNX6 resulted in the downregulation of N-cadherin and ZEB1, a mesenchymal gene and transcriptional factor, respectively, whereas upregulating E-cadherin resulted in the inhibition of the migration and invasive capacity of the cancer cells. Complementing these observations, SNX6 was highly expressed in pancreatic cancer patients and correlated with poor prognosis.
A series of studies published by Bendris et al. revealed that SNX9 is differentially expressed through different stages of breast cancer, which can affect the functions of SNX9 as executed in different cancer cells. However, the exact mechanism regulating this phenomenon remains unclear [116]. It was also reported that SNX9 contributes to the regulation of invadopodia formation and maturation in an inverse manner, as it interacts with RhoA and TKS5, respectively. Additionally, SNX9 mediated the endocytosis of MT1-MMP metalloproteinase, a crucial component for extracellular matrix degradation [117], thereby suggesting a possible role of SNX9 in regulating cancer cell invasiveness.
SNX10 deficiency promotes the initiation and progression of colorectal cancer in mice. Consistent to these observations, SNX10 expression levels were found to be much lower in human colorectal cancer tissues. Further analysis into the mechanism of SNX10 driven colorectal tumorigenesis revealed that loss of SNX10 activates chaperone-mediated autophagy (CMA) and increases the degradation of tumor suppressor p21 in a CMA dependent manner [118]. This was confirmed via in vitro analysis as well as mouse models, which showed an increase in tumor cell proliferation and viability upon SNX10 downregulation, which can be reversed upon exogenously increasing the expression levels of SNX10. Another mechanism underlining SNX10 mediated colorectal cancer progression was suggested through autophagy. SNX10 deficiency disrupts the autophagic degradation of proto-oncogene SRC, which in turn activates the STAT3 pathway, thus promoting colorectal cancer initiation and progression [119]. Interestingly, aberrant expression of SNX10 has been reported in hepatocellular carcinoma. Cervantes-Anaya et al. reported that the mRNA and protein levels of SNX10 are inversely expressed in the tumors of hepatocellular carcinoma which led on to the identification of microRNA-30d which was found to negatively regulate the translation of SNX10 protein levels [120]. Therefore, not only has SNX10 been deemed as having tumor suppressor roles in colorectal cancer, it has also been suggested to be a putative marker for hepatocellular cancer with tumor suppressive potential likely regulated by a microRNA.

6. Discovering SNX Proteins in Liquid Cancers

As we have described above the incidence and roles of SNX proteins in various solid tumors, in this section we will discuss the reports of SNX family members particularly in liquid cancers, such as lymphoma, leukemia, and myeloma (Table 4 provides a summary of known reports identifying SNXs in various cancers). SNX2 was the first member of the SNX family to be reported in acute myeloid leukemia (AML) by the group of Fuchs et al. With the help of screening a human kidney library, this group identified SNX2 as an interacting partner of the formin-binding protein 17 (FBP17), a known MLL-fusion protein found in AML patients [121]. They also observed co-immunoprecipitation of SNX2 with EGFR, which is known to be significantly upregulated in AML [122]. Therefore, this study suggested that SNX2 can alter AML development by regulating the EGFR pathway in a FBP17-MLL dependent manner. Later in 2011, Ernst et al. identified SNX2 as a novel gene fusion partner to ABL1 in B-cell acute lymphoblastic leukemia (ALL) [123]. Follow up research then revealed that the SNX2-ABL1 fusion gene is directly and functionally involved in the pathogenesis of ALL [124]. Additionally, the gene expression profile of ALL cells harboring a SNX2-ABL1 fusion transcript was similar to that of BCR-ABL1, a previously identified prognostic markers for ALL [125]. Furthermore, in vitro analysis using murine Ba/F3 cells showed that SNX2-ABL1 protein expression, similar to BCR-ABL1, transformed the cells such that they were able to proliferate in an IL-3 independent manner [126]. However, when comparing responses to kinase inhibitors, imatinib and dasatinib, it was observed that murine Ba/F3 cells carrying the SNX2-ABL1 fusion were less sensitive to the treatment as the rate of apoptosis was much lower in the SNX2-ABL1 versus BCR-ABL1 expressing cells. Therefore, further research is deemed necessary to identify selective tyrosine kinase inhibitors in response to SNX2-ABL1 specific chimeric fusion protein expressed in ALL patients.
Similarly, a study has implicated a therapeutic potential of SNX10 in treating B-cell non-Hodgkin lymphoma (B-NHL) [127]. This study highlighted the potent anti-tumor activity and sensitivity of apilimod against B-NHL, as displayed in both in vitro as well as in vivo models of the disease. In order to determine the underlying genomic factors that attributes B-NHL cells sensitive towards apilimod, they performed a genome wide screen using CRISPR and identified SNX10, among other candidates, whose loss of function resulted in B-NHL resistance to apilimod. They also observed that lack of SNX10 in B-NHL cells impairs maturation of lysosomes, disrupts autophagy clearance, and promotes activation of transcription factor (TFEB). Overall, this study identified a novel role of SNX10 in B-NHL drug sensitivity towards apilimod [127].
Although direct validation studies have not been conducted yet, there are speculations that SNX9 may also play a role in the development of liquid cancers. A recent siRNA mediated knockdown study has reported that the loss of SNX9 dramatically reduces internalization of ADAM9 [128]. Previous studies have shown that ADAM9 is a pro-tumorigenic protein and promotes aggressive phenotype in several cancers, including myeloma [129]. Additionally, depletion of SNX9 resulted in the increase of ADAM9 cell surface expression levels and its functional activity as indicated by an increase in the shedding of its substrate EphB4. Therefore, this study provides evidence towards the tumor-suppressor roles of SNX9 and also indicates towards its potential in the prevention of liquid cancers, among others, in an ADAM9-dependent manner.
SNX27, the most unique member of the SNX family, has also been identified to play a role in leukemia. An RNAi screen conducted in primary AML cells reported that SNX27 promotes AML tumorigenesis and may serve as a potential prognostic marker, since a lack of SNX27 dramatically reduces tumor cell growth and viability [130]. More recently, it was demonstrated that Tax-1 interacts with SNX27 in a PDZ-dependent manner [131]. Tax1 is a regulatory gene required for the replication and pathogenesis of adult T-cell leukemia causing virus, HTLV-1 [132]. This study showed that Tax-1 overexpression reduces GLUT1 cell surface levels and increases its lysosomal degradation, an effect previously demonstrated by SNX27 loss of function studies [3]. Additionally, GLUT1 is a well-known HTLV-1 receptor molecule previously implicated in viral fusion, shedding, and infectivity [133]. Therefore, this study describes a novel role of SNX27 in the pathogenesis of leukemia causing virus as maintenance of GLUT1 cell surface levels via Tax-1/SNX27 interaction provides novel insight into post-infection regulation of HTLV-1 receptor molecules.
Table 4. Summary of SNXs reported in various cancers.
Table 4. Summary of SNXs reported in various cancers.
MemberType of CancerAssociated ProteinMechanism of ActionRef.
SNX27Acute myeloid leukemiaUnknownSNX27 deletion reduces tumor cell viability and proliferation[130]
Adult T-cell leukemiaTax1Tax-1 interacts with SNX27 to regulate GLUT-1 and pathogenesis of ATL causing virus HTLV-1[131]
Breast cancerMT1-MMPSNX27 directly recycles MT1-MMP and enhances tumor invasiveness[50]
Breast cancerE-cadherin, VimentinSNX27 deletion regulates EMT markers and reduces tumor cell viability and proliferation[87]
SNX1Gastric cancerE-cadherinUpregulation of SNX1 reduces tumor cell
invasion and aggressiveness
[109,110]
SNX5Thyroid cancerUnknownSNX5 expression decreases with tumor progression[111,112]
Head and neck squamous cell carcinomaMcl-1SNX5 deletion induces apoptosis[113]
SNX6Pancreatic cancerZEB-1, E-cadherin, N-cadherinSNX6 deletion regulates EMT markers and
reduces tumor cell aggressiveness
[115]
SNX9Breast cancerMT1-MMPUpregulation of SNX9 promotes MT1-MMP endocytosis and reduces tumor invasiveness[117]
[ADAM9 implicated in leukemia and lymphoma]ADAM9SNX9 negatively regulates ADAM9 surface levels[128]
SNX2Acute myeloid leukemiaFBP17, EGFRSNX2 interacts with prognostic FBP17/MLL fusion protein and regulates EGFR signaling[121]
B-cell acute lymphoblastic leukemiaABL1Expression of SNX2/ABL1 fusion protein reduces drug sensitivity[123]
SNX10B-cell non-Hodgkin lymphomaUnknownDeletion of SNX10 reduces drug sensitivity[127]
Colorectal Cancerp21, SRCOverall downregulated; loss of SNX10 decreases p21 tumor suppressor and increases SRC[118,119]
Hepatocellular carcinomamiR-30dSNX10 is silenced by miR-30d oncomir which promotes cancer progression[120]

7. Future Perspectives and Conclusion

A lot of research has been focused on unravelling the fundamental roles of SNXs in synchronizing the various stages of endosomal cargo selection and transport [2]. The SNX family comprises members that are functionally and structurally distinct from one another, which influences their involvement in endosomal cargo sorting and trafficking mechanisms [13]. Genome wide analysis provides a helpful platform to identify differential expression levels of target SNXs in various cancers; however, identifying the underlying mechanisms driving the correlation between SNXs and cancer remain to be properly investigated. Determining the importance of individual SNX protein in maintaining intracellular protein homeostasis will help to understand the etiology of cancers.
SNX27 is the most unique member of the SNX family of proteins which mediates endocytic recycling of several critical transmembrane proteins in a PDZ-domain dependent manner [1]. However, research is still ongoing to identify and validate SNX27 binding cargoes that may have critical roles in maintaining human health and regulating diseases. Additionally, the non-canonical mechanisms and functions of SNX27 remain unexplored. Therefore, biological and structural analysis into understanding the mechanisms of SNX27-retromer-mediated sorting and transport of endocytosed proteins may provide novel insights into the otherwise unknown role(s) of SNX27.
Several studies have implicated SNX27 in regulating various cancers, particularly breast cancer [87]. This has been supported by genome wide database analysis, in vitro gene regulation studies, as well as in vivo xenograft models [87]. We reported the reduced SNX27 in suppressing autophagy in colonic cancer cell lines [134]. However, in order to fully understand the molecular role of SNX27 in pathways involved in the individual steps of tumorigenesis, detailed research is required that focus on using appropriate and innovative techniques and experimental cancer models to better mimic the tumor microenvironment in vivo. Microarray analysis has helped in the identification of several PDZ-binding motif carrying proteins that are yet to be experimentally validated [88]. Various GPCRs have been implicated in tumorigenesis as well as associations with SNX27, however the exact mechanism driving SNX27 mediated interaction in cancer has not been explored [86]. Therefore, future studies are required to study and demonstrate the crucial role of SNX27 in tumorigenesis. New insights into the roles of SNX27 in various cancers will devise and strategize novel SNX27 associated therapeutic targets for prevention and treatment of these challenging diseases.

Author Contributions

S.D. performed literature search and detailed analyses and summary of related literature, prepared the draft text. J.S. designed the study/project, obtained funds, summarized literature, finalized writing, and directed the project. All authors contributed to the article and approved the submitted version. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the UIC Cancer Center, the NIDDK/National Institutes of Health grant R01DK105118 and R01DK114126, VA Merit Award VA 1 I01 BX004824-01, and DOD CDMRP BC191198 to Jun Sun. The study sponsors played no role in the study design, data collection, analysis, and interpretation of data. The contents do not represent the views of the United States Department of Veterans Affairs or the United States Government.

Conflicts of Interest

The authors declare no conflict of interest. The sponsors had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Temkin, P.; Lauffer, B.; Jäger, S.; Cimermancic, P.; Krogan, N.J.; von Zastrow, M. SNX27 mediates retromer tubule entry and endosome-to-plasma membrane trafficking of signaling receptors. Nat. Cell Biol. 2011, 13, 715–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Teasdale, R.D.; Collins, B.M. Insights into the PX (phox-homology) domain and SNX (sorting nexin) protein families: Structures, functions, and roles in disease. Biochem. J. 2012, 441, 39–59. [Google Scholar] [CrossRef] [PubMed]
  3. Steinberg, F.; Gallon, M.; Winfield, M.; Thomas, E.C.; Bell, A.J.; Heesom, K.J.; Tavaré, J.M.; Cullen, P.J. A global analysis of SNX27-retromer assembly and cargo specificity reveals a function in glucose and metal ion transport. Nat. Cell Biol. 2013, 15, 461–471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Rojas, R.; Kametaka, S.; Haft, C.R.; Bonifacino, J.S. Interchangeable but essential functions of SNX1 and SNX2 in the association of retromer with endosomes and the trafficking of mannose 6-phosphate receptors. Mol. Cell Biol. 2007, 25, 1112–1124. [Google Scholar] [CrossRef] [Green Version]
  5. Yang, Z.; Follett, J.; Kerr, M.C.; Clairfeuille, T.; Chandra, M.; Collins, B.M.; Teasdale, R.D. Sorting nexin 27 (SNX27) regulates the trafficking and activity of the glutamine transporter ASCT2. J. Biol. Chem. 2018, 293, 6802–6811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Sheff, D.R.; Daro, E.A.; Hull, M.; Mellman, I. The receptor recycling pathway contains two distinct populations of early endosomes with different sorting functions. J. Cell Biol. 1999, 145, 123–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Zhang, H.; Huang, T.; Hong, Y.; Yang, W.; Zhang, X.; Luo, H.; Xu, H.; Wang, X. The retromer complex and sorting nexins in neurodegenerative diseases. Front. Aging Neurosci. 2018, 10, 79. [Google Scholar] [CrossRef]
  8. Seaman, M.N.; McCaffery, J.M.; Emr, S.D. A membrane coat complex essential for endosome-to-Golgi retrograde transport in yeast. J. Cell Biol. 1998, 142, 665–681. [Google Scholar] [CrossRef] [Green Version]
  9. Hierro, A.; Rojas, A.L.; Rojas, R.; Murthy, N.; Effantin, G.; Kajava, A.V.; Steven, A.C.; Bonifacino, J.S.; Hurley, J.H. Functional architecture of the retromer cargo-recognition complex. Nature 2007, 449, 1063–1067. [Google Scholar] [CrossRef] [Green Version]
  10. Burd, C.; Cullen, P.J. Retromer: A master conductor of endosome sorting. Cold Spring Harb Perspect Biol. 2014, 6, a016774. [Google Scholar] [CrossRef]
  11. Harrison, M.S.; Hung, C.S.; Liu, T.T.; Christiano, R.; Walther, T.C.; Burd, C.G. A mechanism for retromer endosomal coat complex assembly with cargo. Proc. Natl. Acad. Sci. USA 2014, 111, 267–272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Clague, M.J.; Jones, A.T.; Mills, I.G.; Walker, D.M.; Urbé, S. Regulation of early-endosome dynamics by phosphatidylinositol 3-phosphate binding proteins. Biochem. Soc. Trans. 1999, 27, 662–666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Cullen, P.J. Endosomal sorting and signaling: An emerging role for sorting nexins. Nat. Rev. Mol. Cell Biol. 2008, 9, 574–582. [Google Scholar] [CrossRef] [PubMed]
  14. Xu, Y.; Seet, L.F.; Hanson, B.; Hong, W. The Phox homology (PX) domain, a new player in phosphoinositide signalling. Biochem. J. 2001, 360, 513–530. [Google Scholar] [CrossRef] [PubMed]
  15. Haft, C.R.; de la Luz Sierra, M.; Barr, V.A.; Haft, D.H.; Taylor, S.I. Identification of a family of sorting nexin molecules and characterization of their association with receptors. Mol. Cell Biol. 1998, 18, 7278–7287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Elwell, C.; Engel, J. Emerging role of retromer in modulating pathogen growth. Trends Microbiol. 2018, 26, 769–780. [Google Scholar] [CrossRef]
  17. McNally KE and Cullen, P.J. Endosomal retrieval of cargo: Retromer is not alone. Trends Cell Biol. 2018, 28, 807–822. [Google Scholar] [CrossRef] [Green Version]
  18. Wassmer, T.; Attar, N.; Bujny, M.V.; Oakley, J.; Traer, C.J.; Cullen, P.J. A loss-of-function screen reveals SNX5 and SNX6 as potential components of the mammalian retromer. J. Cell Sci. 2007, 120 Pt 1, 45–54. [Google Scholar] [CrossRef] [Green Version]
  19. Lucas, M.; Gershlick, D.C.; Vidaurrazaga, A.; Rojas, A.L.; Bonifacino, J.S.; Hierro, A. Structural Mechanism for Cargo Recognition by the Retromer Complex. Cell 2016, 167, 1623–1635. [Google Scholar] [CrossRef] [Green Version]
  20. Zhang, P.; Wu, Y.; Belenkaya, T.Y.; Lin, X. SNX3 controls Wingless/Wnt secretion through regulating retromer-dependent recycling of Wntless. Cell Res. 2011, 21, 1677–1690. [Google Scholar] [CrossRef]
  21. Tabuchi, M.; Yanatori, I.; Kawai, Y.; Kishi, F. Retromer-mediated direct sorting is required for proper endosomal recycling of the mammalian iron transporter DMT1. J. Cell Sci. 2010, 123, 756–766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Yong, X.; Zhao, L.; Deng, W.; Sun, H.; Zhou, X.; Mao, L.; Hu, W.; Shen, X.; Sun, Q.; Billadeau, D.D.; et al. Mechanism of cargo recognition by retromer-linked SNX-BAR proteins. PLoS Biol. 2020, 18, e3000631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Kurten, R.C.; Cadena, D.L.; Gill, G.N. Enhanced degradation of EGF receptors by a sorting nexin, SNX1. Science 1996, 272, 1008–1110. [Google Scholar] [CrossRef] [PubMed]
  24. Kvainickas, A.; Jimenez-Orgaz, A.; Nägele, H.; Hu, Z.; Dengjel, J.; Steinberg, F. Cargo-selective SNX-BAR proteins mediate retromer trimer independent retrograde transport. J. Cell Biol. 2017, 216, 3677–3693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ghai, R.; Mobli, M.; Norwood, S.J.; Bugarcic, A.; Teasdale, R.D.; King, G.F.; Collins, B.M. Phox homology band 4.1/ezrin/radixin/moesin-like proteins function as molecular scaffolds that interact with cargo receptors and Ras GTPases. Proc. Natl. Acad. Sci. USA 2011, 108, 7763–7768. [Google Scholar] [CrossRef] [Green Version]
  26. Lee, S.; Chang, J.; Blackstone, C. FAM21 directs SNX27-retromer cargoes to the plasma membrane by preventing transport to the Golgi apparatus. Nat. Commun. 2016, 7, 10939. [Google Scholar] [CrossRef] [Green Version]
  27. Soulet, F.; Yarar, D.; Leonard, M.; Schmid, S.L. SNX9 regulates dynamin assembly and is required for efficient clathrin-mediated endocytosis. Mol. Biol. Cell. 2005, 16, 2058–2067. [Google Scholar] [CrossRef] [Green Version]
  28. Bendris, N.; Schmid, S.L. Endocytosis, metastasis and beyond: Multiple facets of SNX9. Trends Cell Biol. 2017, 27, 189–200. [Google Scholar] [CrossRef] [Green Version]
  29. Fuster, J.J.; González, J.M.; Edo, M.D.; Viana, R.; Boya, P.; Cervera, J.; Verges, M.; Rivera, J.; Andrés, V. Tumor suppressor p27(Kip1) undergoes endolysosomal degradation through its interaction with sorting nexin 6. FASEB J. 2010, 24, 2998–3009. [Google Scholar] [CrossRef]
  30. Li, C.; Ma, W.; Yin, S.; Liang, X.; Shu, X.; Pei, D.; Egan, T.M.; Huang, J.; Pan, A.; Li, Z. Sorting Nexin 11 Regulates Lysosomal Degradation of Plasma Membrane TRPV3. Traffic 2016, 17, 500–514. [Google Scholar] [CrossRef]
  31. Yong, X.; Zhao, L.; Hu, W.; Sun, Q.; Ham, H.; Liu, Z.; Ren, J.; Zhang, Z.; Zhou, Y.; Yang, Q.; et al. SNX27-FERM-SNX1 complex structure rationalizes divergent trafficking pathways by SNX17 and SNX27. Proc. Natl. Acad. Sci. USA 2021, 118, e2105510118. [Google Scholar] [CrossRef] [PubMed]
  32. Joubert, L.; Hanson, B.; Barthet, G.; Sebben, M.; Claeysen, S.; Hong, W.; Marin, P.; Dumuis, A.; Bockaert, J. New sorting nexin (SNX27) and NHERF specifically interact with the 5-HT4a receptor splice variant: Roles in receptor targeting. J. Cell Sci. 2004, 117 Pt 22, 5367–5379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Lunn, M.L.; Nassirpour, R.; Arrabit, C.; Tan, J.; Mcleod, I.; Arias, C.M.; Sawchenko, P.E.; Yates, J.R.; Slesinger, P.A. A unique sorting nexin regulates trafficking of potassium channels via a PDZ domain interaction. Nat. Neurosci. 2007, 10, 1249–1259. [Google Scholar] [CrossRef] [PubMed]
  34. Rincon, E.; Santos, T.; Avila-Flores, A.; Albar, J.P.; Lalioti, V.; Lei, C.; Hong, W.; Merida, I. Proteomics identification of sorting nexin 27 as a diacylglycerol kinase zeta-associated protein: New diacylglycerol kinase roles in endocytic recycling. Mol. Cell Proteom. 2007, 6, 1073–1087. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Clairfeuille, T.; Mas, C.; Chan, A.S.; Yang, Z.; Tello-Lafoz, M.; Chandra, M.; Widagdo, J.; Kerr, M.C.; Paul, B.; Mérida, I.; et al. Molecular code for endosomal recycling of phosphorylated cargos by the SNX27-retromer complex. Nat. Struct Mol. Biol. 2016, 23, 921–932. [Google Scholar] [CrossRef]
  36. Lauffer, B.E.; Melero, C.; Temkin, P.; Lei, C.; Hong, W.; Kortemme, T.; von Zastrow, M. SNX27 mediates PDZ-directed sorting from endosomes to the plasma membrane. J. Cell Biol. 2010, 190, 565–574. [Google Scholar] [CrossRef] [Green Version]
  37. van Weering, J.R.; Sessions, R.B.; Traer, C.J.; Kloer, D.P.; Bhatia, V.K.; Stamou, D.; Carlsson, S.R.; Hurley, J.H.; Cullen, P.J. Molecular basis for SNX-BAR-mediated assembly of distinct endosomal sorting tubules. EMBO J. 2012, 31, 4466–4480. [Google Scholar] [CrossRef] [Green Version]
  38. Derivery, E.; Sousa, C.; Gautier, J.J.; Lombard, B.; Loew, D.; Gautreau, A. The Arp2/3 activator WASH controls the fission of endosomes through a large multiprotein complex. Dev. Cell. 2009, 17, 712–723. [Google Scholar] [CrossRef] [Green Version]
  39. Harbour, M.E.; Breusegem, S.Y.; Seaman, M.N. Recruitment of the endosomal WASH complex is mediated by the extended ‘tail’ of Fam21 binding to the retromer protein Vps35. Biochem. J. 2012, 442, 209–220. [Google Scholar] [CrossRef]
  40. Seaman, M.N.; Gautreau, A.; Billadeau, D.D. Retromer-mediated endosomal protein sorting: All WASHed up! Trends Cell Biol. 2013, 23, 522–528. [Google Scholar] [CrossRef]
  41. Temkin, P.; Morishita, W.; Goswami, D.; Arendt, K.; Chen, L.; Malenka, R. The Retromer Supports AMPA Receptor Trafficking During LTP. Neuron 2017, 94, 74–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Stangl, A.; Elliott, P.R.; Pinto-Fernandez, A.; Bonham, S.; Harrison, L.; Schaub, A.; Kutzner, K.; Keusekotten, K.; Pfluger, P.T.; El Oualid, F.; et al. Regulation of the endosomal SNX27-retromer by OTULIN. Nat. Commun. 2019, 10, 4320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Singh, V.; Yang, J.; Cha, B.; Chen, T.E.; Sarker, R.; Yin, J.; Avula, L.R.; Tse, M.; Donowitz, M. Sorting nexin 27 regulates basal and stimulated brush border trafficking of NHE3. Mol. Biol. Cell. 2015, 26, 2030–2043. [Google Scholar] [CrossRef]
  44. Bannert, K.; Berlin, P.; Reiner, J.; Lemcke, H.; David, R.; Engelmann, R.; Lamprecht, G. SNX27 regulates DRA activity and mediates its direct recycling by PDZ-interaction in early endosomes at the apical pole of Caco2 cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2020, 318, G854–G869. [Google Scholar] [CrossRef] [PubMed]
  45. Balana, B.; Maslennikov, I.; Kwiatkowski, W.; Stern, K.M.; Bahima, L.; Choe, S.; Slesinger, P.A. Mechanism underlying selective regulation of G protein-gated inwardly rectifying potassium channels by the psychostimulant-sensitive sorting nexin 27. Proc. Natl. Acad. Sci. USA 2011, 108, 5831–5836. [Google Scholar] [CrossRef] [Green Version]
  46. Munoz, M.B.; Slesinger, P.A. Sorting nexin 27 regulation of G protein-gated inwardly rectifying K(+) channels attenuates in vivo cocaine response. Neuron 2014, 82, 659–669. [Google Scholar] [CrossRef] [Green Version]
  47. Chan, A.S.; Clairfeuille, T.; Landao-Bassonga, E.; Kinna, G.; Ng, P.Y.; Loo, L.S.; Cheng, T.S.; Zheng, M.; Hong, W.; Teasdale, R.D.; et al. Sorting nexin 27 couples PTHR trafficking to retromer for signal regulation in osteoblasts during bone growth. Mol. Biol. Cell. 2016, 27, 1367–1382. [Google Scholar] [CrossRef]
  48. Lin, T.B.; Lai, C.Y.; Hsieh, M.C.; Wang, H.H.; Cheng, J.K.; Chau, Y.P.; Chen, G.D.; Peng, H.Y. VPS26A-SNX27 interaction-dependent mGluR5 recycling in dorsal horn neurons mediates neuropathic pain in rats. J. Neurosci. 2015, 35, 14943–14955. [Google Scholar] [CrossRef] [Green Version]
  49. Sun, L.; Hu, X.; Chen, W.; He, W.; Zhang, Z.; Wang, T. Sorting nexin 27 interacts with Fzd7 and mediates Wnt signalling. Biosci. Rep. 2016, 36, e00296. [Google Scholar] [CrossRef]
  50. Sharma, P.; Parveen, S.; Shah, L.; Mukherjee, M.; Kalaidzidis, Y.; Kozielski, A.; Rosato, R.; Chang, J.; Datta, S. SNX27–retromer assembly recycles MT1-MMP to invadopodia and promotes breast cancer metastasis. J. Cell Biol. 2020, 219, e201812098. [Google Scholar] [CrossRef]
  51. Zimmerman, S.P.; Hueschen, C.L.; Malide, D.; Milgram, S.L.; Playford, M.P. Sorting nexin 27 (SNX27) associates with zonula occludens-2 (ZO-2) and modulates the epithelial tight junction. Biochem. J. 2013, 455, 95–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Hussain, N.K.; Diering, G.H.; Sole, J.; Anggono, V.; Huganir, R.L. Sorting nexin 27 regulates basal and activity-dependent trafficking of AMPARs. Proc. Natl. Acad. Sci. USA 2014, 111, 11840–11845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Nakagawa, T.; Asahi, M. β1-adrenergic receptor recycles via a membranous organelle, recycling endosome, by binding with sorting nexin27. J. Membr Biol. 2013, 246, 571–579. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bauch, C.; Koliwer, J.; Buck, F.; Hönck, H.H.; Kreienkamp, H.J. Subcellular sorting of the G-protein coupled mouse somatostatin receptor 5 by a network of PDZ-domain containing proteins. PLoS ONE 2014, 9, e88529. [Google Scholar] [CrossRef] [Green Version]
  55. MacNeil, A.J.; Mansour, M.; Pohajdak, B. Sorting nexin 27 interacts with the cytohesin associated scaffolding protein (CASP) in lymphocytes. Biochem. Biophys. Res. Commun. 2007, 359, 848–853. [Google Scholar] [CrossRef] [PubMed]
  56. Cai, L.; Loo, L.S.; Atlashkin, V.; Hanson, B.J.; Hong, W. Deficiency of sorting nexin 27 (SNX27) leads to growth retardation and elevated levels of N-methyl-D-aspartate (NMDA) receptor 2C (NR2C). Mol. Cell Biol. 2011, 31, 1734–1747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Choi, H.J.; Jang, H.J.; Park, E.; Tingskov, S.J.; Nørregaard, R.; Jung, H.J.; Kwon, T.H. Sorting Nexin 27 Regulates the Lysosomal Degradation of Aquaporin-2 Protein in the Kidney Collecting Duct. Cells 2020, 9, 1208. [Google Scholar] [CrossRef] [PubMed]
  58. Valdes, J.L.; Tang, J.; McDermott, M.I.; Kuo, J.C.; Zimmerman, S.P.; Wincovitch, S.M.; Waterman, C.M.; Milgram, S.L.; Playford, M.P. Sorting nexin 27 protein regulates trafficking of a p21-activated kinase [2] interacting exchange factor (β-Pix)-G protein-coupled receptor kinase interacting protein (GIT) complex via a PDZ domain interaction. J. Biol. Chem. 2011, 286, 39403–39416. [Google Scholar] [CrossRef] [Green Version]
  59. Hayashi, H.; Naoi, S.; Nakagawa, T.; Nishikawa, T.; Fukuda, H.; Imajoh-Ohmi, S.; Kondo, A.; Kubo, K.; Yabuki, T.; Hattori, A.; et al. Sorting nexin 27 interacts with multidrug resistance-associated protein 4 (MRP4) and mediates internalization of MRP4. J Biol. Chem. 2012, 287, 15054–15065. [Google Scholar] [CrossRef] [Green Version]
  60. Huang, T.Y.; Zhao, Y.; Li, X.; Wang, X.; Tseng, I.C.; Thompson, R.; Tu, S.; Willnow, T.E.; Zhang, Y.W.; Xu, H. SNX27 and SORLA Interact to Reduce Amyloidogenic Subcellular Distribution and Processing of Amyloid Precursor Protein. J. Neurosci. 2016, 36, 7996–8011. [Google Scholar] [CrossRef]
  61. Meraviglia, V.; Ulivi, A.F.; Boccazzi, M.; Valenza, F.; Fratangeli, A.; Passafaro, M.; Lecca, D.; Stagni, F.; Giacomini, A.; Bartesaghi, R.; et al. SNX27, a protein involved in down syndrome, regulates GRP17 trafficking and oligodendrocyte differentiation. Glia 2016, 64, 1437–1460. [Google Scholar] [CrossRef] [PubMed]
  62. Nourry, C.; Grant, S.G.N.; and Borg, J.P. PDZ domain proteins: Plug and play! Sci. STKE 2003, 179, re7. [Google Scholar] [CrossRef] [PubMed]
  63. Damseh, N.; Danson, C.M.; Al-Ashhab, M.; Abu-Libdeh, B.; Gallon, M.; Sharma, K.; Yaacov, B.; Coulthard, E.; Caldwell, M.A.; Edvardson, S.; et al. A defect in the retromer accessory protein, SNX27, manifests by infantile myoclonic epilepsy and neurodegeneration. Neurogenetics 2015, 16, 215–221. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Tello-Lafoz, M.; Ghai, R.; Collins, B.; Mérida, I. A role for novel lipid interactions in the dynamic recruitment of SNX27 to the T-cell immune synapse. Bioarchitecture 2014, 4, 215–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Gunzer, M.; Weishaupt, C.; Hillmer, A.; Basoglu, Y.; Friedl, P.; Dittmar, K.E.; Kolanus, W.; Varga, G.; Grabbe, S. A spectrum of biophysical interaction modes between T cells and different antigen-presenting cells during priming in 3-D collagen and in vivo. Blood 2004, 104, 2801–2809. [Google Scholar] [CrossRef] [Green Version]
  66. Onnis, A.; Finetti, F.; Baldari, C.T. Vesicular Trafficking to the Immune Synapse: How to Assemble Receptor-Tailored Pathways from a Basic Building Set. Front. Immunol. 2016, 7, 50. [Google Scholar] [CrossRef] [Green Version]
  67. Monks, C.R.F.; Freiberg, B.A.; Kupfer, H.; Sciaky, N.; Kupfer, A. Three-dimensional segregation of supramolecular activation clusters in T cells. Nature 1998, 395, 82–86. [Google Scholar] [CrossRef]
  68. Monjas, A.; Alcover, A.; Alarcón, B. Engaged and bystander T cell receptors are down-modulated by different endocytotic pathways. J. Biol. Chem. 2004, 279, 55376–55384. [Google Scholar] [CrossRef] [Green Version]
  69. Soares, H.; Lasserre, R.; Alcover, A. Orchestrating cytoskeleton and intracellular vesicle traffic to build functional immunological synapses. Immunol. Rev. 2013, 256, 118–132. [Google Scholar] [CrossRef]
  70. Rincon, E.; de Guinoa, J.S.; Gharbi, S.I.; Sorzano, C.O.S.; Carrasco, Y.R.; Merida, I. Translocation dynamics of sorting nexin 27 in activated T cells. J. Cell Sci. 2011, 124, 776–788. [Google Scholar] [CrossRef]
  71. Ghai, R.; Tello-Lafoz, M.; Norwood, S.J.; Yang, Z.; Clairfeuille, T.; Teasdale, R.D.; Merida, I.; Collins, B.M. Phosphoinositide binding by the SNX27 FERM domain regulates its localization at the immune synapse of activated T-cells. J. Cell Sci. 2015, 128, 553–565. [Google Scholar] [PubMed] [Green Version]
  72. Tello-Lafoz, M.; Martínez-Martínez, G.; Rodríguez-Rodríguez, C.; Albar, J.P.; Huse, M.; Gharbi, S.; Merida, I. Sorting nexin 27 interactome in T-lymphocytes identifies zona occludens-2 dynamic redistribution at the immune synapse. Traffic 2017, 18, 491–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Almena, M.; Mérida, I. Shaping up the membrane: Diacylglycerol coordinates spatial orientation of signaling. Trends Biochem. Sci. 2011, 36, 593–603. [Google Scholar] [CrossRef] [PubMed]
  74. Ávila-Flores, A.; Arranz-Nicolás, J.; Andrada, E.; Soutar, D.; Mérida, I. Predominant contribution of DGKζ over DGKα in the control of PKC/PDK-1-regulated functions in T cells. Immunol. Cell Biol. 2017, 95, 549–563. [Google Scholar] [CrossRef] [PubMed]
  75. Tello-Lafoz, M.; Rodríguez-Rodríguez, C.; Kinna, G.; Loo, L.S.; Hong, W.; Collins, B.M.; Teasdale, R.D.; Mérida, I. SNX27 links DGKζ to the control of transcriptional and metabolic programs in T lymphocytes. Sci. Rep. 2017, 7, 16361. [Google Scholar] [CrossRef] [Green Version]
  76. McMillan, K.J.; Korswagen, H.C.; Cullen, P.J. The emerging role of retromer in neuroprotection. Curr. Opin. Cell Biol. 2017, 47, 72–82. [Google Scholar] [CrossRef] [Green Version]
  77. Wang, X.; Zhao, Y.; Zhang, X.; Badie, H.; Zhou, Y.; Mu, Y.; Loo, L.S.; Cai, L.; Thompson, R.C.; Yang, B.; et al. Loss of sorting nexin 27 contributes to excitatory synaptic dysfunction by modulating glutamate receptor recycling in Down’s syndrome. Nat. Med. 2013, 19, 473–480. [Google Scholar] [CrossRef] [Green Version]
  78. Loo, L.S.; Tang, N.; Al-Haddawi, M.; Dawe, G.S.; Hong, W. A role for sorting nexin 27 in AMPA receptor trafficking. Nat. Commun. 2014, 5, 3176. [Google Scholar] [CrossRef] [Green Version]
  79. Fujiyama, K.; Kajii, Y.; Hiraoka, S.; Nishikawa, T. Differential regulation by stimulants of neocortical expression of mrt1, arc, and homer1a mRNA in the rats treated with repeated methamphetamine. Synapse 2003, 49, 143–149. [Google Scholar] [CrossRef]
  80. Willén, K.; Sroka, A.; Takahashi, R.H.; Gouras, G.K. Heterogeneous Association of Alzheimer’s Disease-Linked Amyloid-β and Amyloid-β Protein Precursor with Synapses. J. Alzheimers Dis. 2017, 60, 511–524. [Google Scholar] [CrossRef]
  81. Cataldo, A.; Rebeck, G.W.; Ghetri, B.; Hulette, C.; Lippa, C.; Van Broeckhoven, C.; van Duijn, C.; Cras, P.; Bogdanovic, N.; Bird, T.; et al. Endocytic disturbances distinguish among subtypes of Alzheimer’s disease and related disorders. Ann. Neurol. 2001, 50, 661–665. [Google Scholar] [CrossRef] [PubMed]
  82. Wang, X.; Huang, T.; Zhao, Y.; Zheng, Q.; Thompson, R.C.; Bu, G.; Zhang, Y.W.; Hong, W.; Xu, H. Sorting nexin 27 regulates Aβ production through modulating γ-secretase activity. Cell Rep. 2014, 9, 1023–1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Nielsen, M.S.; Gustafsen, C.; Madsen, P.; Nyengaard, J.R.; Hermey, G.; Bakke, O.; Mari, M.; Schu, P.; Pohlmann, R.; Dennes, A.; et al. Sorting by the cytoplasmic domain of the amyloid precursor protein binding receptor SorLA. Mol. Cell Biol. 2007, 27, 6842–6851. [Google Scholar] [CrossRef] [Green Version]
  84. Olmos-Serrano, J.L.; Kang, H.J.; Tyler, W.A.; Silbereis, J.C.; Cheng, F.; Zhu, Y.; Pletikos, M.; Jankovic-Rapan, L.; Cramer, N.P.; Galdzicki, Z.; et al. Down Syndrome Developmental Brain Transcriptome Reveals Defective Oligodendrocyte Differentiation and Myelination. Neuron 2016, 89, 1208–1222. [Google Scholar] [CrossRef] [Green Version]
  85. Niciu, M.J.; Kelmendi, B.; Sanacora, G. Overview of glutamatergic neurotransmission in the nervous system. Pharmacol. Biochem. Behav. 2012, 100, 656–664. [Google Scholar] [CrossRef] [Green Version]
  86. Bao, Z.; Zhou, S.; Zhou, H. Sorting Nexin 27 as a potential target in G protein coupled receptor recycling for cancer therapy. Oncol. Rep. 2020, 44, 1779–1786. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, J.; Li, K.; Zhang, Y.; Lu, R.; Wu, S.; Tang, J.; Xia, Y.; Sun, J. Deletion of sorting nexin 27 suppresses proliferation in highly aggressive breast cancer MDA-MB-231 cells in vitro and in vivo. BMC Cancer 2019, 19, 555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Lee, H.J.; Zheng, J.J. PDZ domains and their binding partners: Structure, specificity, and modification. Cell Commun. Signal 2010, 8, 8. [Google Scholar] [CrossRef] [Green Version]
  89. Du, J.; Li, X.H.; Li, Y.J. Glutamate in peripheral organs: Biology and pharmacology. Eur. J. Pharmacol. 2016, 784, 42–48. [Google Scholar] [CrossRef]
  90. Liu, B.; Zhao, S.; Qi, C.; Zhao, X.; Liu, B.; Hao, F.; Zhao, Z. Inhibition of metabotropic glutamate receptor 5 facilitates hypoxia-induced glioma cell death. Brain Res. 2019, 1704, 241–248. [Google Scholar] [CrossRef]
  91. Takanami, I.; Inoue, Y.; Gika, M. G-protein inwardly rectifying potassium channel 1 (GIRK 1) gene expression correlates with tumor progression in non-small cell lung cancer. BMC Cancer 2004, 4, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Liu, H.C.; Wang, C.; Xie, N.; Zhuang, Z.; Liu, X.; Hou, J.; Huang, H. Activation of adrenergic receptor β2 promotes tumor progression and epithelial mesenchymal transition in tongue squamous cell carcinoma. Int. J. Mol. Med. 2018, 41, 147–154. [Google Scholar] [CrossRef] [PubMed]
  93. Calvo, N.; Martin, M.J.; de Boland, A.R.; Gentili, C. Involvement of ERK1/2, p38 MAPK, and PI3K/Akt signaling pathways in the regulation of cell cycle progression by PTHrP in colon adenocarcinoma cells. Biochem. Cell Biol. 2014, 92, 305–315. [Google Scholar] [CrossRef] [PubMed]
  94. Zeng, C.M.; Chen, Z.; Fu, L. Frizzled receptors as potential therapeutic targets in human cancers. Int. J. Mol. Sci. 2018, 19, 1543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Cole, S.W.; Sood, A.K. Molecular pathways: Beta-adrenergic signaling in cancer. Clin. Cancer Res. 2012, 18, 1201–1206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Lupp, A.; Klenk, C.; Röcken, C.; Evert, M.; Mawrin, C.; Schulz, S. Immunohistochemical identification of the PTHR1 parathyroid hormone receptor in normal and neoplastic human tissues. Eur. J. Endocrinol. 2010, 162, 979–986. [Google Scholar] [CrossRef] [Green Version]
  97. Prickett, T.D.; Samuels, Y. Molecular pathways: Dysregulated glutamatergic signaling pathways in cancer. Clin. Cancer Res. 2012, 18, 4240–4246. [Google Scholar] [CrossRef] [Green Version]
  98. Zhang, Z.; Liu, R.; Shuai, Y.; Huang, Y.; Jin, R.; Wang, X.; Luo, J. ASCT2 (SLC1A5)-dependent glutamine uptake is involved in the progression of head and neck squamous cell carcinoma. Br. J. Cancer 2020, 122, 82–93. [Google Scholar] [CrossRef]
  99. Sahores, A.; Carozzo, A.; May, M.; Gomez, N.; Di Siervi, N.; De Sousa Serro, M.; Yaneff, A. Multidrug transporter MRP4/ABCC4 as a key determinant of pancreatic cancer aggressiveness. Sci. Rep. 2020, 10, 14217. [Google Scholar] [CrossRef]
  100. Barbieri, F.; Bajetto, A.; Pattarozzi, A.; Gatti, M.; Würth, R.; Thellung, S.; Corsaro, A.; Villa, V.; Nizzari, M.; Florio, T. Peptide receptor targeting in cancer: The somatostatin paradigm. Int. J. Pept. 2013, 2013, 926295. [Google Scholar] [CrossRef]
  101. Hong, W.; Gu, Y.; Guan, R.; Xie, D.; Zhou, H.; Yu, M. Pan-cancer analysis of the CASP gene family in relation to survival, tumor-infiltrating immune cells and therapeutic targets. Genomics 2020, 112, 4304–4315. [Google Scholar] [CrossRef] [PubMed]
  102. North, W.G.; Liu, F.; Lin, L.Z.; Tian, R.; Akerman, B. NMDA receptors are important regulators of pancreatic cancer and are potential targets for treatment. Clin. Pharmacol. 2017, 9, 79–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Li, M.; He, M.; Xu, F.; Guan, Y.; Tian, J.; Wan, Z.; Zhou, H.; Gao, M.; Chong, T. Abnormal expression and the significant prognostic value of aquaporins in clear cell renal cell carcinoma. PLoS ONE 2022, 17, e0264553. [Google Scholar] [CrossRef] [PubMed]
  104. Keum, S.; Yang, S.J.; Park, E.; Kang, T.; Choi, J.H.; Jeong, J.; Hwang, Y.E.; Kim, J.W.; Park, D.; Rhee, S. Beta-Pix-dynamin 2 complex promotes colorectal cancer progression by facilitating membrane dynamics. Cell Oncol. 2021, 44, 1287–1305. [Google Scholar] [CrossRef] [PubMed]
  105. Zou, Z.; Tao, T.; Li, H.; Zhu, X. mTOR signaling pathway and mTOR inhibitors in cancer: Progress and challenges. Cell Biosci. 2020, 10, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Carvalho, K.C.; Cunha, I.W.; Rocha, R.M.; Ayala, F.R.; Cajaíba, M.M.; Begnami, M.D.; Vilela, R.S.; Paiva, G.R.; Andrade, R.G.; Soares, F.A. GLUT1 expression in malignant tumors and its use as an immunodiagnostic marker. Clinics 2011, 66, 965–972. [Google Scholar] [CrossRef] [Green Version]
  107. Riese, M.J.; Moon, E.K.; Johnson, B.D.; Albelda, S.M. Diacylglycerol Kinases (DGKs): Novel Targets for Improving T Cell Activity in Cancer. Front. Cell Dev. Biol. 2016, 4, 108. [Google Scholar] [CrossRef] [Green Version]
  108. Cullen, P.J.; Korswagen, H.C. Sorting nexins provide diversity for retromer-dependent trafficking events. Nat. Cell Biol. 2011, 14, 29–37. [Google Scholar] [CrossRef]
  109. Zhan, X.Y.; Zhang, Y.; Zhai, E.; Zhu, Q.Y.; He, Y. Sorting nexin-1 is a candidate tumor suppressor and potential prognostic marker in gastric cancer. PeerJ 2018, 29, e4829. [Google Scholar] [CrossRef] [Green Version]
  110. Bryant, D.M.; Kerr, M.C.; Hammond, L.A.; Joseph, S.R.; Mostov, K.E.; Teasdale, R.D.; Stow, J.L. EGF induces macropinocytosis and SNX1-modulated recycling of E-cadherin. J. Cell Sci. 2007, 120 Pt 10, 1818–1828. [Google Scholar] [CrossRef]
  111. Ara, S.; Kikuchi, T.; Matsumiya, H.; Kojima, T.; Kubo, T.; Ye, R.C.; Sato, A.; Kon, S.I.; Honma, T.; Asakura, K.; et al. Sorting nexin 5 of a new diagnostic marker of papillary thyroid carcinoma regulates Caspase-2. Cancer Sci. 2012, 103, 1356–1362. [Google Scholar] [CrossRef] [PubMed]
  112. Jitsukawa, S.; Kamekura, R.; Kawata, K.; Ito, F.; Sato, A.; Matsumiya, H.; Nagaya, T.; Yamashita, K.; Kubo, T.; Kikuchi, T.; et al. Loss of sorting nexin 5 stabilizes internalized growth factor receptors to promote thyroid cancer progression. J. Pathol. 2017, 243, 342–353. [Google Scholar] [CrossRef] [PubMed]
  113. Cai, J.; Sun, M.; Hu, B.; Windle, B.; Ge, X.; Li, G.; Sun, Y. Sorting Nexin 5 Controls Head and Neck Squamous Cell Carcinoma Progression by Modulating FBW7. J. Cancer 2019, 10, 2942–2952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. He, L.; Torres-Lockhart, K.; Forster, N.; Ramakrishnan, S.; Greninger, P.; Garnett, M.J.; McDermott, U.; Rothenberg, S.M.; Benes, C.H.; Ellisen, L.W. Mcl-1 and FBW7 control a dominant survival pathway underlying HDAC and Bcl-2 inhibitor synergy in squamous cell carcinoma. Cancer Discov. 2013, 3, 324–337. [Google Scholar] [CrossRef] [Green Version]
  115. Hu, P.; Liang, Y.; Hu, Q.; Wang, H.; Cai, Z.; He, J.; Cai, J.; Liu, M.; Qin, Y.; Yu, X.; et al. SNX6 predicts poor prognosis and contributes to the metastasis of pancreatic cancer cells via activating epithelial-mesenchymal transition. Acta Biochim. Biophys. Sin. 2018, 50, 1075–1084. [Google Scholar] [CrossRef] [Green Version]
  116. Bendris, N.; Stearns, C.J.; Reis, C.R.; Rodriguez-Canales, J.; Liu, H.; Witkiewicz, A.W.; Schmid, S.L. Sorting nexin 9 negatively regulates invadopodia formation and function in cancer cells. J. Cell Sci. 2016, 129, 2804–2816. [Google Scholar] [CrossRef] [Green Version]
  117. Linder, S. The matrix corroded: Podosomes and invadopodia in extracellular matrix degradation. Trends Cell Biol. 2007, 17, 107–117. [Google Scholar] [CrossRef]
  118. Zhang, S.; Hu, B.; You, Y.; Yang, Z.; Liu, L.; Tang, H.; Bao, W.; Guan, Y.; Shen, X. Sorting nexin 10 acts as a tumor suppressor in tumorigenesis and progression of colorectal cancer through regulating chaperone mediated autophagy degradation of p21Cip1/WAF1. Cancer Lett. 2018, 419, 116–127. [Google Scholar] [CrossRef]
  119. Zhang, S.; Yang, Z.; Bao, W.; Liu, L.; You, Y.; Wang, X.; Shao, L.; Fu, W.; Kou, X.; Shen, W.; et al. SNX10 (sorting nexin 10) inhibits colorectal cancer initiation and progression by controlling autophagic degradation of SRC. Autophagy 2020, 16, 735–749. [Google Scholar] [CrossRef]
  120. Cervantes-Anaya, N.; Ponciano-Gómez, A.; López-Álvarez, G.S.; Gonzalez-Reyes, C.; Hernández-Garcia, S.; Cabañas-Cortes, M.A.; Garrido-Guerrero, J.E.; Villa-Treviño, S. Downregulation of sorting nexin 10 is associated with overexpression of miR-30d during liver cancer progression in rats. Tumour. Biol. 2017, 39, 1010428317695932. [Google Scholar] [CrossRef]
  121. Fuchs, U.; Rehkamp, G.; Haas, O.A.; Slany, R.; Kōnig, M.; Bojesen, S.; Bohle, R.M.; Damm-Welk, C.; Ludwig, W.D.; Harbott, J.; et al. The human formin-binding protein 17 (FBP17) interacts with sorting nexin, SNX2, and is an MLL-fusion partner in acute myelogeneous leukemia. Proc. Natl. Acad. Sci. USA 2001, 98, 8756–8761. [Google Scholar] [CrossRef] [Green Version]
  122. Mahmud, H.; Kornblau, S.M.; Ter Elst, A.; Scherpen, F.J.; Qiu, Y.H.; Coombes, K.R.; de Bont, E.S. Epidermal growth factor receptor is expressed and active in a subset of acute myeloid leukemia. J. Hematol. Oncol. 2016, 9, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Ernst, T.; Score, J.; Deininger, M.; Hidalgo-Curtis, C.; Lackie, P.; Ershler, W.B.; Goldman, J.M.; Cross, N.C.; Grand, F. Identification of FOXP1 and SNX2 as novel ABL1 fusion partners in acute lymphoblastic leukaemia. Br. J. Haematol. 2011, 153, 43–46. [Google Scholar] [CrossRef] [PubMed]
  124. Mu, Q.; Guo, L.; Hu, Y.; Sheng, L.; Zhang, Y.; Wu, N.; Chen, Y.; Shi, C.; Shi, S.; Wu, Y.; et al. SNX2-ABL1-positive acute lymphoblastic leukemia possibly has a poor prognosis. Leuk. Lymphoma. 2017, 58, 1–3. [Google Scholar] [CrossRef] [PubMed]
  125. Masuzawa, A.; Kiyotani, C.; Osumi, T.; Shioda, Y.; Iijima, K.; Tomita, O.; Nakabayashi, K.; Oboki, K.; Yasuda, K.; Sakamoto, H.; et al. Poor responses to tyrosine kinase inhibitors in a child with precursor B-cell acute lymphoblastic leukemia with SNX2-ABL1 chimeric transcript. Eur. J. Haematol. 2014, 92, 263–267. [Google Scholar] [CrossRef]
  126. Tomita, O.; Iijima, K.; Ishibashi, T.; Osumi, T.; Kobayashi, K.; Okita, H.; Saito, M.; Mori, T.; Shimizu, T.; Kiyokawa, N. Sensitivity of SNX2-ABL1 toward tyrosine kinase inhibitors distinct from that of BCR-ABL1. Leuk. Res. 2014, 38, 361–370. [Google Scholar] [CrossRef]
  127. Gayle, S.; Landrette, S.; Beeharry, N.; Conrad, C.; Hernandez, M.; Beckett, P.; Ferguson, S.M.; Mandelkern, T.; Zheng, M.; Xu, T.; et al. Identification of apilimod as a first-in-class PIKfyve kinase inhibitor for treatment of B-cell non-Hodgkin lymphoma. Blood 2017, 129, 1768–1778. [Google Scholar] [CrossRef]
  128. Mygind, K.J.; Störiko, T.; Freiberg, M.L.; Samsøe-Petersen, J.; Schwarz, J.; Andersen, O.M.; Kveiborg, M. Sorting nexin 9 (SNX9) regulates levels of the transmembrane ADAM9 at the cell surface. J. Biol. Chem. 2018, 293, 8077–8088. [Google Scholar] [CrossRef] [Green Version]
  129. Karadag, A.; Zhou, M.; Croucher, P.I. ADAM-9 (MDC-9/meltrin-gamma), a member of the a disintegrin and metalloproteinase family, regulates myeloma-cell-induced interleukin-6 production in osteoblasts by direct interaction with the alpha(v)beta5 integrin. Blood 2006, 107, 3271–3278. [Google Scholar] [CrossRef] [Green Version]
  130. Wermke, M.; Camgoz, A.; Paszkowski-Rogacz, M.; Thieme, S.; von Bonin, M.; Dahl, A.; Platzbecker, U.; Theis, M.; Ehninger, G.; Brenner, S.; et al. RNAi profiling of primary human AML cells identifies ROCK1 as a therapeutic target and nominates fasudil as an antileukemic drug. Blood 2015, 125, 3760–3768. [Google Scholar] [CrossRef]
  131. Al-Saleem, J.; Dirksen, W.P.; Martinez, M.P.; Shkriabai, N.; Kvaratskhelia, M.; Ratner, L.; Green, P.L. HTLV-1 Tax-1 interacts with SNX27 to regulate cellular localization of the HTLV-1 receptor molecule, GLUT1. PLoS ONE 2019, 14, e0214059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Akagi, T.; Ono, H.; Shimotohno, K. Characterization of T cells immortalized by Tax1 of human T-cell leukemia virus type 1. Blood 1995, 86, 4243–4249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Jones, K.S.; Lambert, S.; Bouttier, M.; Bénit, L.; Ruscetti, F.W.; Hermine, O.; Pique, C. Molecular aspects of HTLV-1 entry: Functional domains of the HTLV-1 surface subunit (SU) and their relationships to the entry receptors. Viruses 2011, 3, 794–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Li, K.; Wu, S.; Zhang, Y.; Playford, M.P.; Sun, J. Depletion of Endosome-Associated Protein SNX27 Inhibits Autophage in Human Colonic Cancer Cells. Gastroenterology 2015, 148, S952. [Google Scholar] [CrossRef]
Figure 1. Structure and functional domains of SNX27.
Figure 1. Structure and functional domains of SNX27.
Cancers 15 00070 g001
Table 1. Characteristic features of different SNX domains.
Table 1. Characteristic features of different SNX domains.
SubfamilyMembersPrimary RolesLinks to Processes Indicated in Health and Disease Regulation
SNX-PXSNX3Assists in the recruitment of the retromer complex to early endosomesSNX3-retromer complex distinctly carries out cargo recognition,
SNX10binding, and retrograde recycling to the TGN and
SNX11SNX3 is also important for neurite development
SNX12
SNX16SNX10 regulates autophagy
SNX20
SNX21SNX11 mediates lysosomal degradation of EGFR and TRPV3
SNX22
SNX24SNX16 regulates endosomal membrane dynamics and mediates
SNX29trafficking between early and late endosomes
SNX-BARSNX1Additional C-terminal BAR domain comprising three α- helicesSNX9 and SNX18 carry an additional SH3 domain and
SNX2regulate clathrin-mediated endocytosis of cargoes
SNX4
SNX5SNX1 mediates trafficking of mGluR1 in hippocampus and
SNX6SNX1 SNPs are found in brains of Alzheimer’s patients
SNX7Heterodimer of SNX1:SNX2 and SNX5:SNX6 is involved in retromer mediated trafficking
SNX8SNX2 is overexpressed in the hypothalamus of aging mice
SNX9
SNX18SNX4, 6, 7, 8 regulate Aβ generation by mediating APP and
SNX30BACE1 trafficking and lysosomal degradation
SNX32Involved in membrane tubulation
SNX33SNX5 is associated with trafficking of Dopamine receptor 1
SNX-FERMSNX17 SNX27 SNX31C-terminal FERM domain assists in endocytic traffickingand lysosomal degradationSNX17 and 31 target cargoes carrying a NPxY/NxxY motif and
cause lysosomal degradation in a FERM-dependent manner
SNX27-FERM domain binds with SNX1/2 homodimer to form
SNX27 carries an additional N-terminal PDZ domain that helps to sort and recycle proteins to the cell surfacethe SNX27/SNX-BAR/Retromer complex
SNX27 modulates synaptic plasticity in the brain and is
associated with Epilepsy, Alzheimer’s, and Down’s Syndrome
Table 2. Identified binding partners of SNX27.
Table 2. Identified binding partners of SNX27.
Target IDTarget NameRef.
β2-arβ2 adrenergic receptor[1]
5-HT4.a/R5-hydroxytryptamine type 4 receptor[32]
GIRK2G protein-gated inwardly rectifying potassium 2[45]
GIRK3G protein-gated inwardly rectifying potassium 3[46]
PTHRParathyroid hormone 1 receptor[47]
mGluR5Metabotropic glutamate receptor 5[48]
FZD7Frizzled receptor 7[49]
GLUT1Glucose transporter 1[3]
ATP7AATPase copper transporting alpha[3]
ASCT2Alanine-, serine-, cysteine-preferring transporter 2[5]
RasRas GTPase[25]
OTULINOTU Deubiquitinase With Linear Linkage Specificity[42]
NHE3Sodium (Na+)/hydrogen (H+) exchanger 3[43]
DRADownregulated in adenoma[44]
MT1-MMPMembrane type 1 matrix metalloproteinase[50]
DGKζDiacylglycerol kinase zeta[34]
ZO-2Zonula occludens 2[51]
AMPAα-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor[52]
β1-arβ1 adrenergic receptor [53]
SSTR5mouse Somatostatin receptor subtype 5[54]
CASPCytohesin associated scaffolding protein[55]
NR2CN-methyl-D-aspartate (NMDA) receptor 2C[56]
AQP2Aquaporin 2[57]
β-Pixβ-PAK-interacting exchange factor[58]
GitG-protein receptor kinase interacting target[58]
MRP4Multidrug resistance-associated protein 4[59]
SorLASorting-related receptor with A-type repeats[60]
GRP17Gadd related protein, 17 kDa[61]
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

Deb, S.; Sun, J. Endosomal Sorting Protein SNX27 and Its Emerging Roles in Human Cancers. Cancers 2023, 15, 70. https://doi.org/10.3390/cancers15010070

AMA Style

Deb S, Sun J. Endosomal Sorting Protein SNX27 and Its Emerging Roles in Human Cancers. Cancers. 2023; 15(1):70. https://doi.org/10.3390/cancers15010070

Chicago/Turabian Style

Deb, Shreya, and Jun Sun. 2023. "Endosomal Sorting Protein SNX27 and Its Emerging Roles in Human Cancers" Cancers 15, no. 1: 70. https://doi.org/10.3390/cancers15010070

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