Next Article in Journal
Dialysis as a Novel Adjuvant Treatment for Malignant Cancers
Next Article in Special Issue
Inhibition of LIFR Blocks Adiposity-Driven Endometrioid Endometrial Cancer Growth
Previous Article in Journal
Mechanical Properties of Colorectal Cancer Cells Determined by Dynamic Atomic Force Microscopy: A Novel Biomarker
Previous Article in Special Issue
Metformin as a Potential Treatment Option for Endometriosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Atypical Macropinocytosis Contributes to Malignant Progression: A Review of Recent Evidence in Endometrioid Endometrial Cancer Cells

Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Japan
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(20), 5056; https://doi.org/10.3390/cancers14205056
Submission received: 5 September 2022 / Revised: 1 October 2022 / Accepted: 13 October 2022 / Published: 15 October 2022

Abstract

:

Simple Summary

A novel type of macropinocytosis has been identified as a trigger for the malignant progression of endometrial cancer. Transiently reducing epithelial barrier homeostasis leads to macropinocytosis by splitting between adjacent cells in endometrioid endometrial cancer. Macropinocytosis causes morphological changes in well-differentiated to poorly differentiated cancer cells. Inhibition of macropinocytosis promotes a persistent dormant state in the intrinsic KRAS-mutated cancer cell line Sawano. This review focuses on the mechanisms of atypical macropinocytosis and its effects on cellular function, and it describes the physiological processes involved in inducing resting conditions in endometrioid endometrial cancer cells.

Abstract

Macropinocytosis is an essential mechanism for the non-specific uptake of extracellular fluids and solutes. In recent years, additional functions have been identified in macropinocytosis, such as the intracellular introduction pathway of drugs, bacterial and viral infection pathways, and nutritional supplement pathway of cancer cells. However, little is known about the changes in cell function after macropinocytosis. Recently, it has been reported that macropinocytosis is essential for endometrial cancer cells to initiate malignant progression in a dormant state. Macropinocytosis is formed by a temporary split of adjacent bicellular junctions of epithelial sheets, rather than from the apical surface or basal membrane, as a result of the transient reduction of tight junction homeostasis. This novel type of macropinocytosis has been suggested to be associated with the malignant pathology of endometriosis and endometrioid endometrial carcinoma. This review outlines the induction of malignant progression of endometrial cancer cells by macropinocytosis based on a new mechanism and the potential preventive mechanism of its malignant progression.

Graphical Abstract

1. Introduction

Macropinocytosis has long been known to be one of the mechanisms involved in the uptake of extracellular components. This phenomenon was discovered in macrophages in 1931 and is a widely confirmed biological phenomenon from amoebas and slime molds to mammalian cells [1]. Endocytosis is the mechanism by which cells incorporate extracellular factors. Endocytosis is an essential mechanism for the uptake of the extracellular components required for housekeeping tasks. According to the typical definition, they are classified into two types: phagocytosis, which involves the uptake of solid substances, and pinocytosis, which involves the uptake of fluid components [2]. Macropinocytosis is a process that nonspecifically incorporates extracellular components on a large scale. Macropinocytosis is a physiologically important function of immune cells, endothelial cells, and epithelial cells. In cancer cells, high macropinocytosis activity is used as a supply pathway of amino acids required for cell proliferation [3]. A recent study identified a new role for macropinocytosis, inducing the initiation of malignant progression of endometrial cancer cells [4]. Macropinocytosis is attributed to the disruption of epithelial cell homeostasis. Evidence that macropinocytosis contributes to the malignant process may point toward new targets for the prevention of cancer.

2. Uptake Mechanisms of Extracellular Components

Most phagocytic cells actively incorporate the target factors (Figure 1) [5]. Professional phagocytes, such as macrophages, neutrophils, and dendritic cells, and non-professional phagocytes, such as fibroblasts and epithelial cells, incorporate targets, which include solid components containing apoptotic cells, through receptor-mediated endocytosis [6]. Professional phagocytes efficiently engulf extracellular components through complement-mediated opsonization. Phagosome formation involves membrane ruffling via PI3K-mediated actin reorganization [7]. The coordinated reconstruction of cell membranes is also required [8]. Mycobacterium tuberculosis uses phagocytosis as a route of entry into the cell, but its mechanism is different from that of foreign body removal to prevent its own degradation [9]. Some cancer cells express CD47 [10]. The interaction of CD47 with the SIRPα of macrophages renders cancer cells resistant to phagocytosis, thereby promoting tumor growth and metastasis [11]. Endometrial cancer also expresses CD47, and it has been pointed out that its inhibition may be a therapeutic target [12].
Pinocytosis can be classified according to the size of vesicles formed [13]. Small vesicle-mediated pinocytosis is further classified into the following three types: clathrin-mediated, caveolae pathways and those mediated by neither. In the clathrin-mediated pathway, clathrin-based triskelion vesicles facilitate intracellular transport [14]. Caveolae form vesicles associated with caveolin-1, which are smaller than clathrin-coated vesicles [15]. These small vesicles originate from concavity of the cell membrane [16]. Pinocytosis, accompanied by the formation of large vesicles, is called macropinocytosis. Macropinocytosis does not require coat proteins such as clathrin for uptake. In addition, it is believed that there is no communication mechanism or interaction between the incorporated target and the cell [17]. In macropinocytosis, extracellular factors are incorporated into membrane protrusions via a pathway that involves extensive actin rearrangement [18] and membrane ruffling [19]. A mathematical model has been developed to explain the mechanism of formation of a large macropinocytic cup during macropinocytosis [20].
The extracellular uptake mechanism is used as a route for viral infection [21]. Phagocytosis allows herpes simplex viruses (HSV-1) and acanthamoeba polyphaga mimivirus (AMPV) to enter cells, whereas in clathrin-mediated endocytosis, human immunodeficiency virus type 1 (HIV-1) and influenza A virus (IAV) enter cells, while simian virus (SV40) and BK virus enter via caveolae. Adenovirus 3 (Ad3), coxsackievirus B (CVB), and respiratory syncytial virus (RSV) enter cells through macropinocytosis [21,22,23].

3. A Universal Role of Macropinocytosis

Among immune system cells, macrophages and dendritic cells constantly perform macropinocytosis to eliminate foreign substances and present antigens [24]. B cells, CD4+ T cells, and CD8+ T cells utilize macropinocytosis for their physiological functions [25,26].
In fibroblasts, growth factors, such as PDGF and EGF, stimulate Ras and Src via activation of RTKs, leading to macropinocytosis [27,28]. Circular dorsal ruffles (CDRs) occur on the cell surface and are involved in the formation of macropinocytic cups [29]. The PI3K-Akt-TSC1-Rheb-mTORC1 pathway is associated with growth factor-dependent macropinosome formation [30]. The macropinosome fuses with the lysosome, and its contents are absorbed into the cell [3]. mTORC1/2 complexes regulate the recycling of essential biomolecules in lysosomes through interactions with the nutrient source sensor TSC1/2 [31,32].
In endothelial cells, macropinocytosis is a pathway for growth factor receptor internalization in a growth factor-dependent manner. Cell surface VEGFR2 is incorporated via macropinocytosis by Cdc42 activation upon VEGF stimulation [33]. Cell surface FGFR1 is incorporated into Rab5-positive endosomes by macropinocytosis via RhoG activation upon FGF2 stimulation [34]. Macropinocytosis is required for the activation of intracellular signaling pathways that are dependent on growth factor stimulation.
Since collective epithelial cells have established cell polarity, they incorporate extracellular components from the apical and basal membranes. In polar MDCK cells, uptake occurs via clathrin-mediated and clathrin-independent pathways on the apical surface, whereas caveolae-mediated uptake occurs on the basal membrane [13]. Two types of plasma membrane ruffles precede macropinocytosis: circular dorsal ruffles (CDRs) arising from the apical cell surface and planar ruffles arising from the cell periphery [3]. These processes involve the activation of the RhoG-Rac pathway and reorganization of actin filaments [35]. Recently, it was reported that macropinocytosis is necessary for breaking out contact inhibition and initiating stratified cell growth in well-differentiated human endometrial cancer cells (Figure 1) [4]. While macropinocytosis, which is Rac-dependent, occurs through the temporary split of adjacent cell junctions; there is no protrusion of the cell membrane prior to macropinocytosis. The triggering of this macropinocytosis is based on a transient reduction in epithelial homeostasis by ligand stimulation of the tight junction-associated protein lipolysis-stimulated lipoprotein receptor (LSR).

4. Disruption of Epithelial Homeostasis Triggers Cancer Malignancy

4.1. Regulation of Epithelial Homeostasis by Cell Polarity

To maintain homeostasis in epithelial cells, it is necessary to establish an adhesion apparatus between adjacent cells and concomitantly establish cell polarity [36]. Adherence junction formation precedes cell–cell junction formation [37]. Adherence junctions interact with the actin cytoskeleton through the nectin-afadin and cadherin-α/β-catenin pathways [38]. The actin cytoskeleton forms circumferential actomyosin bundles and increases the cell thickness [39]. Shroom3 and Lulu1/2 activate circumferential actomyosin bundles via ROCK [40,41]. Tight junctions are established by polar proteins PAR3/PAR6/aPKC, which interact with circumferential actomyosin bundles via ZO1/2/3 [42]. In mature epithelial cell sheets with sufficient epithelial barrier function, occludin (OCLN), claudins (CLDNs), and JAM-A are localized in the tight junction region between adjacent cells, whereas tricellulin (Tric) accumulates in the tricellular contact region in an LSR dependent manner. The LSR is required for the localization of Tric to the tricellular contact region [43], but the mechanism by which LSR accumulates in this region remains unclear. OCLN, CLDNs, and Tric are transmembrane proteins with N- and C-termini in the cytoplasm, whereas JAM-A and LSR are single-transmembrane proteins with one or more immunoglobulin-like domains in the extracellular N-terminal region [44]. The LSR is a receptor for apolipoprotein B (ApoB) and ApoE [45], which are associated with lipid metabolism in hepatocytes [46]. LSR localization is dependent on the cell density in epithelial cells. During the establishment of epithelial homeostasis, LSRs are distributed broadly in the bicellular junction region and gradually accumulate in the tricellular contact region [47]. This change in localization is reversible. LSR withdrawal from the tricellular contact region impairs epithelial homeostasis [48]. Disruption of epithelial barrier function abolishes cell polarity and promotes dedifferentiation [44,49].

4.2. Regulation of Epithelial Homeostasis by Cell Density

The cell adhesion apparatus senses cell density [50]. Epithelial cells have a contact inhibition mechanism, which is one of the mechanisms that maintains cell density [51,52]. Contact inhibition involves two mechanisms: growth and motility inhibition [53,54]. These contribute to the regulation of cell number during normal development and wound healing [55]. Physical cell loss and carcinogenesis disrupt contact inhibition [56,57]. Loss of contact inhibition and disruption of cell polarity are typical features of the malignant transformation of cancer cells. These molecular mechanisms involve PAR3, TGF-β, and Hippo pathways [53,58]. KRAS mutations are closely associated with the growth of malignant cells [59]. KRAS mutations are typical oncogenes found in one-quarter of adenocarcinomas [60]. KRAS mutations have been found in endometrial carcinomas and adenomyosis [61,62].
Sawano cell was established by endometrial adenocarcinoma of uterus [63]. Sawano has a KRAS G13D heterozygous mutation and wild-type BRaf [4]. According to this study, Sawano cells have high planar motility at low cell densities, whereas at high cell densities, their growth and motility are temporarily arrested due to contact inhibition. After a certain period of time under high cell density conditions, Sawano initiated multilayered cell growth. In addition, in the presence of MAPK inhibitors, the cells maintained a highly differentiated state. Thus, Sawano cells are a good model for analyzing the malignant transformation process of endometrial carcinoma.

5. LSR-Related Molecular Mechanisms Leading to Disruption of Endometrial Homeostasis

5.1. Disruption of Endometrial Homeostasis by TGF-β Signaling

TGF-β is associated with increased migration and malignant transformation of endometrial carcinomas through activation of estrogen-related receptor alpha (ERRα); it is also associated with invasion and epithelial-to-mesenchymal transition (EMT) [64]. TGF-β increases HMGB1 expression and induces changes in LSR localization and downregulation, resulting in reduced epithelial barrier homeostasis [65,66,67,68,69,70,71,72]. TGF-β levels are also elevated in high-grade stages of endometriosis [73]. TGF-β is released from endometrial epithelial cells, endometrial stromal cells, endometriotic mesenchymal stem cells, and peritoneal mesothelial cells and regulates endometriosis pathogenesis [74].

5.2. Disruption of Endometrial Homeostasis by Hippo-Yap Pathway

The Hippo-YAP pathway is involved in the regulation of cell number and apico-basal polarity. This pathway is involved in tight junctions, adherens junctions, the planar cell polarity (PCP) pathway, the mechanotransduction pathway, and growth factor signaling [52]. YAP also regulates the cell size [75]. YAP is localized in the nucleus, resulting in the activation of transcription factors in various cell types, whereas YAP is degraded in the cytoplasm, resulting in transcriptional repression in smaller and columnar cells [76]. In endometriotic tissue, YAP is localized in the cytoplasm, whereas in G1, G2, and G3 stages of endometrioid carcinomas, YAP is localized in the nucleus [77,78]. LSR knockdown enhanced cell motility and invasion in Sawano cells, and YAP knockdown resulted in cell quiescence [77]. Dobutamine, a β-adrenergic receptor agonist, decreases nuclear YAP levels and increases cytoplasmic phosphorylated YAP levels in human U2OS cells [79]. Dobutamine has also been reported to suppress the high expression of YAP in gastric carcinoma, resulting in suppression of cell motility and invasion [80]. In LSR-knockdown Sawano cells, dobutamine treatment suppressed the enhancement of cell motility and invasion, associated with an increase in the amount of phosphorylated YAP [77]. Activation of the transcription factor TEAD by YAP increased the expression of genes associated with cell proliferation and survival [81]. LSR-knockdown Sawano cells induce expression of the Hippo pathway transcription factor TEAD and inducible gene amphiregulin (AREG) [77]. Immunohistochemical staining analysis using tissues from endometriosis and endometrial cancer revealed that the expression of AREG, an EGFR ligand, is observed in the cytoplasm of cells forming endometriotic tissue and that its expression level increases with increasing cancer stage. In Sawano cells, cell motility and invasion, both of which were increased by LSR knockdown, were significantly suppressed by AREG knockdown. Increased expression of AREG has also been observed in the human pancreatic cancer cell line HPAC, wherein LSR is knocked down, and in poorly differentiated pancreatic ductal adenocarcinomas (PDAC), wherein no LSR is localized in the junctional region [66]. Thus, the downregulation of epithelial barrier function caused by the suppression of LSR expression is involved in the regulation of cell motility and invasion through TEAD-dependent AREG expression via the YAP-Hippo pathway.

6. Pathophysiological Changes in Endometriosis and Endometrial Carcinoma Are Accompanied by Changes in LSR Localization

The endometrium is a regenerative tissue that undergoes periodic proliferation and differentiation under the influence of estrogen, progesterone, growth factors, and various cytokines. Menstrual cycle influences the expression and localization of both bicellular and tricellular tight junction proteins. Bicellular tight junction proteins such as CLDN-1, -3, -4, and -7 and OCLN accumulate in the subapical region during the proliferative phase of the endometrium, whereas they are distributed in the basolateral region during the secretory phase [82]. Levels of tricellular tight junction proteins also change dynamically. Both Tric and LSR accumulate in the subapical region during the proliferative phase of the endometrium, wherein Tric is localized in the subapical region during the secretory phase and LSR is widely distributed from the subapical region to the basolateral region [83].
A downregulation of expression of OCLN and CLDN-3, -4, and -7 has been observed in endometriosis [84], and an upregulation of expression of CLDN-3 and -4 has been reported in endometrial cancer [85]. The expression of tricellular tight junction proteins is altered during endometrial pathogenesis [77,83]. These studies demonstrate that the localization of LSR to tricellular contact regions is required for the maintenance of robust endometrial epithelial barrier function. For example, in endometriosis, Tric accumulates in the subapical region, as in the normal human endometrium, whereas LSR is widely distributed not only in the subapical region, but also in the lateral region. In endometrial carcinoma G1, in which most of the gland-like tissue remains, both Tric and LSR are heterogeneously distributed from the subapical to the lateral regions. In stages G2 and G3, the expression levels of both are decreased. Among endometrial cancer-derived cell lines, Sawano, HHUA, and JHMUE-1 cells, which exhibit high to moderate differentiation, express LSR and Tric, whereas spindle-shaped and fibroblast-like JHMUE-2 cells rarely express these markers. Knockdown of LSR in Sawano cells resulted in decreased epithelial barrier function and increased cell motility, invasion, and proliferation, as described above.
In the gland-like structure region of endometrial cancer tissue, upregulation of LSR expression and downregulation of CLDN-1 expression have been observed by immunohistochemical staining analysis [86]. By contrast, in the invasive front area, LSR expression is downregulated and CLDN-1 expression is markedly upregulated. The report has also shown that, CLDN-1 expression upregulates in LSR knocked-down Sawano cells, while the expression levels of OCLN, CLDN-3, -4, and -7 barely change. Prior to LSR knockdown, CLDN-1 is localized in the subapical region, but after the knockdown, it is widely distributed from the subapical region to the basolateral region. Thus, there is a reciprocal relationship between the expression of LSR and CLDN-1, yet almost no regulatory mechanism has been identified. Binding of Sp1 to the CLDN-1 promoter region is required for the regulation of CLDN-1 expression in intestinal epithelium-derived cells [87]. Sp1-binding sites exist in the promoter region of CLDN-1, -4, and -19 [88,89,90]. Sp1 is also involved in the upregulation of CLDN-1 expression in Sawano cells [86]. Sp1 contributes to the expression of matrix metalloproteinases (MMPs) in Sawano cells.
In oral squamous cell carcinoma (OSC) cells expressing CLDN-1, cell invasion is enhanced through cleavage of laminin-5γ2 chains by increased activity of MMP-14 (MT-MMP-1) and MMP-2 [91]. Human colorectal cancer SW480 expressing CLDN-1 have been reported to enhance cell invasion through the activation of MMP-2 and -9 [92]. The initial stage of cell invasion requires reconstitution of the extracellular matrix composition, in addition to the depletion of intercellular junctions [93]. To date, 24 MMPs have been identified [94]. MMP expression varies with the menstrual cycle [95]. MMP-3, -7, -9, and-13 are associated with disease susceptibility in endometriosis [96]. TGF-β is involved in the enhancement of MMP-2 and -3 secretion [97]. LSR knockdown increased the expression levels of MMP-2, -9, -10, and -14 in Sawano cells [86]. MMP-14 is an initiator that activates the MMP cascade by activation of proMMP-2 [98]. In Sawano cells, double knockdown of LSR and CLDN-1 suppresses cell invasion enhanced by LSR knockdown [86]. While some studies have linked increased expression of TGF-β with CLDN-1 expression [99], there exist conflicting reports [100].

7. Internalization Pathways of Tight Junction Proteins

Tight junction complexes form static rigid seals, whereas their components exhibit dynamic behavior. The exclusion of tight junction proteins from the cell surface membrane is associated with internalization by clathrin-, caveolae-, and raft-mediated pathways, as well as by macropinocytosis [101]. In intestinal epithelial cell T84, tight junction proteins OCLN, CLDN-1, and JAM-A are internalized via the IFN-γ-induced macropinocytosis-like pathway [102]. In contrast, in T84 cells, Escherichia coli cytotoxic necrotizing factor-1 (CNF-1) induces caveolae-dependent intracellular translocation of OCLN without affecting the localization of the AJ proteins E-cadherin and β-catenin [103]. In depolymerized MDCK cells, in which actin filamentation is inhibited by latrunculin A, OCLN is internalized via the caveolae [104]. GFP-CLDN-3 was incorporated from the lateral region by vesicles that incorporated the plasma membrane of two adjacent cells [105]. In Caco-2 cells, Tric, OCLN, CLDN-4, and ZO-1 are excluded from tight junctions in a poly L-arginine-dependent manner. However, the underlying mechanism remains unknown [106]. In bovine retinal endothelial cells (BRECs), OCLN is phosphorylated and ubiquitinated 30 min after VEGF stimulation and translocates from tight junctions to early and late endosomes [107]. LSR is excluded from tricellular tight junctions by LSR ligand stimulation, and LSR subsequently colocalizes with Rab5-positive vesicles at bicellular junctions [4]. The pathway by which the LSR translocates from the bicellular region to the intracellular region and the mechanism by which the intracellularly translocated LSR is recycled to the junction region is unclear.

8. Regulation of Cell Motility by Macropinocytosis Induced by LSR Ligand

As mentioned above, decreased expression levels and altered localization of LSR, which accumulates in tricellular tight junctions, alter the localization of Tric, leading to a decrease in the robustness of the epithelial barrier function. Many of these findings were obtained through LSR knockdown experiments, in which it is difficult to temporally control the suppression of gene expression. Hence, Kondoh et al. established an experimental system that enables the simultaneous reduction of LSR expression in all areas of cultured cells using LSR ligands [108]. Administration of the LSR ligand to Sawano cells induces LSR localized in tricellular tight junctions into Rab5-positive intracellular vesicles via bicellular junctions [4]. Under these conditions, the LSR ligand transiently reduced epithelial barrier function, but it had little effect on OCLN expression and localization and did not completely disrupt epithelial barrier function. TEM analysis revealed the presence of tight junctions, adherens junctions, and desmosomes in bicellular junctions. Macropinocytosis does not disrupt the tight junction barrier in a blood-tumor barrier model in rat gliomas [109].
The LSR ligand is a partial C-terminal peptide of the Clostridium perfringens iota-toxin b subunit. LSR bound to C. perfringens iota toxin translocates intracellularly through membrane rafts [110]. LSR expressed in mouse vascular endothelial cells is involved in the establishment of barrier function [111]. Administration of the LSR ligand to mice transiently reduces the endothelial cell barrier function of the blood–brain barrier (BBB) and enhances drug entry into the brain [112]. In the human cancer cell lines Sawano and HPAC, administration of the LSR ligand decreased LSR expression and induced a reduction in barrier function, resulting in enhanced malignant transformation of cancer cells [66,113]. In contact-inhibited monolayers of Sawano cells under high cell density conditions, administration of the LSR ligand resulted in transient and rapid enhancement of planar cell motility [4]. This process requires macropinocytosis, initiated by the cleavage of the gap between adjacent cells (Figure 2). LSR ligand-dependent pinocytosis does not occur in apical or basal membranes. Additionally, no significant accumulation of actin filaments was observed during macropinosome formation. In the presence of the Rac inhibitor NSC23766 or in cells expressing a dominant negative Rac, LSR ligand-dependent macropinocytosis is inhibited. Rac activity is required for JNK activation and is involved in the subsequent enhancement of cell motility. Rac activates JNK [114]. JNK is associated with the regulation of cell motility in various cancer cells [115]. In Sawano cells, LSR ligand stimulation enhances cell motility through JNK activation [113]. JNK inhibitors or knockdown of JNK not only suppresses cell motility but also inhibits macropinocytosis [4]. JNK-mTOR pathway is associated with macropinocytosis in HeLa cells [116].
The mechanism by which cell motility is enhanced following macropinocytosis in DCs has been elucidated. In mature DCs that have undergone macropinocytosis, decreased mTORC1 activity induces the expression of the lysosomal calcium channel TRPML1 and an increase in calcium ions, followed by enhanced cell motility via the activation of myosin II [117]. Myosin II and ROCK play a central role in cell motility associated with actomyosin reorganization [118]. Between neighboring adjacent junctions of Sawano cells, myosin phosphatase MYPT1 accumulates during the contact inhibition phase and phosphorylated myosin accumulates during the cell motility phase [47]. The myosin II inhibitor blebbistatin and the ROCK inhibitor Y27632 did not affect the formation of macropinocytosis, suggesting that LSR ligand-induced macropinocytosis triggers the initiation of planar cell motility.

9. SLC9A1-Mediated Macropinocytosis and Its Associated Regulation of Cancer Malignancy

The amiloride derivative EIPA targets SLC9A1 and suppresses macropinocytosis [119]. SLC9A family of proteins are involved in maintaining intracellular pH homeostasis [120]. These proteins were formerly named as sodium hydrogen exchanger (NHE) family proteins [121]. EIPA inhibits the activation of Rac and Cdc42 by acidifying the intracellular pH and inhibiting macropinocytosis by preventing the translocation of Arp3 to the plasma membrane [122]. The Arp2/3 complex is involved not only in actin filament branching and protrusion but also in phagocytic cup formation [6,123]. During RSV infection, extracellular budding and spread of RSV are based on the regulation of the Arp2/3 complex by transcribed viral proteins [124].
SLC9A1 is a ubiquitously expressed plasma membrane protein that directly interacts with ERM proteins [125] and ROCK [126] in its C-terminal region. SLC9A1 is a scaffold protein for Raf and ERK1/2 [127]. In triple-negative breast cancer MDA-MB-231 cells, SLC9A1 activation triggers malignant transformation [128]. SLC9A1 is involved in collective cell migration in head and neck squamous cell carcinoma [129]. In A431 cells, an increase in intracellular pH due to SLC9A1 activation is required for EGF-dependent macropinocytosis [122]. SLC9A1 has attracted attention as a novel anticancer drug target [130].
In the presence of EIPA, similar to the knockdown of SLC9A1, few macropinosomes were formed upon treatment with the LSR ligand; furthermore, no cell motility was confirmed under these conditions (Figure 3) [4]. Cryo-EM analysis showed that C. perfringens iota toxin, the origin of the LSR ligand, enters the cell under acidic conditions [131]. Transient acidification of the extracellular region by SLC9A1 activation may affect the infection [132]. If the C. perfringens infection pathway is similar to the binding of the LSR ligand to the LSR, it is possible that the infection may be involved in the pathway by which genetically mutated KRAS cells awaken from dormancy. KRAS mutations are representative gene mutations in endometrial cancer [133]. As in endometriosis, lesions of adenomyosis and their surrounding normal tissues have KRAS mutations, even though they are not cancerous [62]. LSR-targeted bacterial infections and associated macropinocytosis may be involved in the malignant transformation of these non-cancerous cells. It has been reported that bicellular tight junction proteins, such as OCLN, CLDNs, and JAM-A, are receptors for adenovirus, papillomavirus, rotavirus, influenza virus, severe acute respiratory syndrome (SARS), and HIV-1 [134]. Understanding the mechanisms by which these exogenous factors are incorporated into cells may help avoid infection and carcinogenesis.
Ras mutations, other than the KRAS G13D mutation, are also associated with macropinocytosis. Analysis of human pancreatic adenocarcinoma-derived MIA PaCa-2 cells with KRAS G12C mutation, human bladder carcinoma-derived T24 cells with HRas G12V, and exogenous KRAS G12V-expressing NIH3T3 cells revealed that these cells obtain glutamine needed for proliferation by uptake of albumin from the extracellular fluid via macropinocytosis [135]. In knock-in mice transfected with KRAS G12D into the pancreas, amino acids necessary for cancer cell growth are acquired through macropinocytosis, a mechanism that is rarely observed in normal cells [136]. These findings implicate macropinocytosis between epithelial barrier homeostasis and initiation of malignant transformation in KRAS-mutant cells. Therefore, it is suggested that the epithelial barrier function may negatively regulate the signaling pathways from mutant KRAS in the dormant state Sawano cells, in which LSR is localized in tricellular tight junctions. It is speculated that LSR may be involved in the initiation of the malignant transformation of endometriosis and endometrial cancer.

10. Conclusions

Atypical macropinocytosis, which is derived from the partial cleavage between adjacent cells, is essential for the initiation of malignant progression of endometrial cancer cells. Aberrant cell proliferation, disruption of differentiation, and EMT are induced by the activation of oncogenes, including Ras, and the inactivation of tumor suppressor genes, including PTEN and p53. These processes involve reorganization of intercellular junctions. LSR may be an initiating factor in the disruption of epithelial barrier homeostasis. Accumulation of LSR in tricellular tight junctions is necessary to maintain the robustness of epithelial barrier function. As shown in Figure 3, changes in the localization of LSR in tricellular tight junctions induce atypical macropinocytosis. Furthermore, macropinocytosis triggers a rapid increase in the motility of endometrial cancer cells and induction of multilayered cell growth. However, it is unclear when and where macropinocytosis occurs in proliferating cancer cells. It is still unclear whether endogenous LSR ligands are involved in the pathological process of endometriosis and the malignant progression of endometrial cancer. The elucidation of these mechanisms may contribute to the development of new therapeutic methods for these diseases. It is expected that mechanisms underlying the formation of atypical macropinocytosis will be discovered.

Author Contributions

Conceptualization, T.K. (Takayuki Kohno) and T.K. (Takashi Kojima); writing—original draft preparation, T.K. (Takayuki Kohno); writing—review and editing, T.K. (Takayuki Kohno) and T.K. (Takashi Kojima); visualization, T.K. (Takayuki Kohno); supervision; T.K. (Takayuki Kohno). All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by JSPS KAKENHI Grant Number JP22K05357 and by The Akiyama Life Science Foundation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. King, J.S.; Kay, R.R. The Origins and Evolution of Macropinocytosis. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2019, 374, 20180158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hoeller, O.; Bolourani, P.; Clark, J.; Stephens, L.R.; Hawkins, P.T.; Weiner, O.D.; Weeks, G.; Kay, R.R. Two Distinct Functions for PI3-Kinases in Macropinocytosis. J. Cell Sci. 2013, 126, 4296–4307. [Google Scholar] [CrossRef] [Green Version]
  3. Recouvreux, M.V.; Commisso, C. Macropinocytosis: A Metabolic Adaptation to Nutrient Stress in Cancer. Front. Endocrinol. 2017, 8, 261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kohno, T.; Konno, T.; Kikuchi, S.; Kondoh, M.; Kojima, T. Translocation of LSR from Tricellular Corners Causes Macropinocytosis at Cell-Cell Interface as a Trigger for Breaking out of Contact Inhibition. FASEB J. 2021, 35, e21742. [Google Scholar] [CrossRef] [PubMed]
  5. Feng, M.; Jiang, W.; Kim, B.Y.S.; Zhang, C.C.; Fu, Y.-X.; Weissman, I.L. Phagocytosis Checkpoints as New Targets for Cancer Immunotherapy. Nat. Rev. Cancer 2019, 19, 568–586. [Google Scholar] [CrossRef]
  6. Uribe-Querol, E.; Rosales, C. Phagocytosis: Our Current Understanding of a Universal Biological Process. Front. Immunol. 2020, 11, 1066. [Google Scholar] [CrossRef] [PubMed]
  7. Schlam, D.; Bagshaw, R.D.; Freeman, S.A.; Collins, R.F.; Pawson, T.; Fairn, G.D.; Grinstein, S. Phosphoinositide 3-Kinase Enables Phagocytosis of Large Particles by Terminating Actin Assembly through Rac/Cdc42 GTPase-Activating Proteins. Nat. Commun. 2015, 6, 8623. [Google Scholar] [CrossRef] [Green Version]
  8. Bohdanowicz, M.; Grinstein, S. Role of Phospholipids in Endocytosis, Phagocytosis, and Macropinocytosis. Physiol. Rev. 2013, 93, 69–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Russell, D.G. Mycobacterium Tuberculosis: Here Today, and Here Tomorrow. Nat. Rev. Mol. Cell Biol. 2001, 2, 569–578. [Google Scholar] [CrossRef] [PubMed]
  10. Matlung, H.L.; Szilagyi, K.; Barclay, N.A.; van den Berg, T.K. The CD47-SIRPα Signaling Axis as an Innate Immune Checkpoint in Cancer. Immunol. Rev. 2017, 276, 145–164. [Google Scholar] [CrossRef] [PubMed]
  11. Logtenberg, M.E.W.; Scheeren, F.A.; Schumacher, T.N. The CD47-SIRPα Immune Checkpoint. Immunity 2020, 52, 742–752. [Google Scholar] [CrossRef] [PubMed]
  12. Gu, S.; Ni, T.; Wang, J.; Liu, Y.; Fan, Q.; Wang, Y.; Huang, T.; Chu, Y.; Sun, X.; Wang, Y. CD47 Blockade Inhibits Tumor Progression through Promoting Phagocytosis of Tumor Cells by M2 Polarized Macrophages in Endometrial Cancer. J. Immunol. Res. 2018, 2018, 6156757. [Google Scholar] [CrossRef] [PubMed]
  13. Sandvig, K.; Kavaliauskiene, S.; Skotland, T. Clathrin-Independent Endocytosis: An Increasing Degree of Complexity. Histochem. Cell Biol. 2018, 150, 107–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Young, A. Structural Insights into the Clathrin Coat. Semin. Cell Dev. Biol. 2007, 18, 448–458. [Google Scholar] [CrossRef] [PubMed]
  15. Rothberg, K.G.; Heuser, J.E.; Donzell, W.C.; Ying, Y.-S.; Glenney, J.R.; Anderson, R.G.W. Caveolin, a Protein Component of Caveolae Membrane Coats. Cell 1992, 68, 673–682. [Google Scholar] [CrossRef]
  16. Rennick, J.J.; Johnston, A.P.R.; Parton, R.G. Key Principles and Methods for Studying the Endocytosis of Biological and Nanoparticle Therapeutics. Nat. Nanotechnol. 2021, 16, 266–276. [Google Scholar] [CrossRef] [PubMed]
  17. Kerr, M.C.; Teasdale, R.D. Defining Macropinocytosis. Traffic 2009, 10, 364–371. [Google Scholar] [CrossRef] [PubMed]
  18. Lin, X.P.; Mintern, J.D.; Gleeson, P.A. Macropinocytosis in Different Cell Types: Similarities and Differences. Membranes 2020, 10, 177. [Google Scholar] [CrossRef] [PubMed]
  19. Buckley, C.M.; King, J.S. Drinking Problems: Mechanisms of Macropinosome Formation and Maturation. FEBS J. 2017, 284, 3778–3790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Saito, N.; Sawai, S. Three-Dimensional Morphodynamic Simulations of Macropinocytic Cups. iScience 2021, 24, 103087. [Google Scholar] [CrossRef] [PubMed]
  21. Mercer, J.; Helenius, A. Virus Entry by Macropinocytosis. Nat. Cell Biol. 2009, 11, 510–520. [Google Scholar] [CrossRef] [PubMed]
  22. Lim, J.P.; Gleeson, P.A. Macropinocytosis: An Endocytic Pathway for Internalising Large Gulps. Immunol. Cell Biol. 2011, 89, 836–843. [Google Scholar] [CrossRef] [PubMed]
  23. Paluck, A.; Osan, J.; Hollingsworth, L.; Talukdar, S.N.; Saegh, A.A.; Mehedi, M. Role of ARP2/3 Complex-Driven Actin Polymerization in RSV Infection. Pathogens 2021, 11, 26. [Google Scholar] [CrossRef] [PubMed]
  24. Norbury, C.C. Drinking a Lot Is Good for Dendritic Cells. Immunology 2006, 117, 443–451. [Google Scholar] [CrossRef] [PubMed]
  25. García-Pérez, B.E.; de la Cruz-López, J.J.; Castañeda-Sánchez, J.I.; Muñóz-Duarte, A.R.; Hernández-Pérez, A.D.; Villegas-Castrejón, H.; García-Latorre, E.; Caamal-Ley, A.; Luna-Herrera, J. Macropinocytosis Is Responsible for the Uptake of Pathogenic and Non-Pathogenic Mycobacteria by B Lymphocytes (Raji Cells). BMC Microbiol. 2012, 12, 246. [Google Scholar] [CrossRef] [Green Version]
  26. Charpentier, J.C.; Chen, D.; Lapinski, P.E.; Turner, J.; Grigorova, I.; Swanson, J.A.; King, P.D. Macropinocytosis Drives T Cell Growth by Sustaining the Activation of MTORC1. Nat. Commun. 2020, 11, 180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Gu, Z.; Noss, E.H.; Hsu, V.W.; Brenner, M.B. Integrins Traffic Rapidly via Circular Dorsal Ruffles and Macropinocytosis during Stimulated Cell Migration. J. Cell Biol. 2011, 193, 61–70. [Google Scholar] [CrossRef]
  28. Mettlen, M.; Platek, A.; van der Smissen, P.; Carpentier, S.; Amyere, M.; Lanzetti, L.; de Diesbach, P.; Tyteca, D.; Courtoy, P.J. Src Triggers Circular Ruffling and Macropinocytosis at the Apical Surface of Polarized MDCK Cells. Traffic 2006, 7, 589–603. [Google Scholar] [CrossRef] [PubMed]
  29. Yoshida, S.; Pacitto, R.; Sesi, C.; Kotula, L.; Swanson, J.A. Dorsal Ruffles Enhance Activation of Akt by Growth Factors. J. Cell Sci. 2018, 131, jcs220517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Yoshida, S.; Pacitto, R.; Inoki, K.; Swanson, J. Macropinocytosis, MTORC1 and Cellular Growth Control. Cell. Mol. Life Sci. 2018, 75, 1227–1239. [Google Scholar] [CrossRef] [PubMed]
  31. Inoki, K.; Zhu, T.; Guan, K.-L.L. TSC2 Mediates Cellular Energy Response to Control Cell Growth and Survival. Cell 2003, 115, 577–590. [Google Scholar] [CrossRef] [Green Version]
  32. Condon, K.J.; Sabatini, D.M. Nutrient Regulation of MTORC1 at a Glance. J. Cell Sci. 2019, 132, jcs222570. [Google Scholar] [CrossRef] [PubMed]
  33. Basagiannis, D.; Zografou, S.; Murphy, C.; Fotsis, T.; Morbidelli, L.; Ziche, M.; Bleck, C.; Mercer, J.; Christoforidis, S. VEGF Induces Signalling and Angiogenesis by Directing VEGFR2 Internalisation through Macropinocytosis. J. Cell Sci. 2016, 129, 4091–4104. [Google Scholar] [CrossRef] [Green Version]
  34. Elfenbein, A.; Lanahan, A.; Zhou, T.X.; Yamasaki, A.; Tkachenko, E.; Matsuda, M.; Simons, M. Syndecan 4 Regulates FGFR1 Signaling in Endothelial Cells by Directing Macropinocytosis. Sci. Signal 2012, 5, ra36. [Google Scholar] [CrossRef] [Green Version]
  35. Valdivia, A.; Goicoechea, S.M.; Awadia, S.; Zinn, A.; Garcia-Mata, R. Regulation of Circular Dorsal Ruffles, Macropinocytosis, and Cell Migration by RhoG and Its Exchange Factor, Trio. Mol. Biol. Cell 2017, 28, 1768–1781. [Google Scholar] [CrossRef] [PubMed]
  36. Zihni, C.; Balda, M.S.; Matter, K. Signalling at Tight Junctions during Epithelial Differentiation and Microbial Pathogenesis. J. Cell Sci. 2014, 127, 3401–3413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Takai, Y.; Nakanishi, H. Nectin and Afadin: Novel Organizers of Intercellular Junctions. J. Cell Sci. 2003, 116, 17–27. [Google Scholar] [CrossRef] [Green Version]
  38. Zihni, C.; Mills, C.; Matter, K.; Balda, M.S. Tight Junctions: From Simple Barriers to Multifunctional Molecular Gates. Nat. Rev. Mol. Cell Biol. 2016, 17, 564–580. [Google Scholar] [CrossRef]
  39. Citi, S. The Mechanobiology of Tight Junctions. Biophys. Rev. 2019, 11, 783–793. [Google Scholar] [CrossRef]
  40. Nishimura, T.; Takeichi, M. Shroom3-Mediated Recruitment of Rho Kinases to the Apical Cell Junctions Regulates Epithelial and Neuroepithelial Planar Remodeling. Development 2008, 135, 1493–1502. [Google Scholar] [CrossRef] [PubMed]
  41. Chu, C.W.; Gerstenzang, E.; Ossipova, O.; Sokol, S.Y. Lulu Regulates Shroom-Induced Apical Constriction during Neural Tube Closure. PLoS ONE 2013, 8, e81854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hurd, T.W.; Gao, L.; Roh, M.H.; Macara, I.G.; Margolis, B. Direct Interaction of Two Polarity Complexes Implicated in Epthelial Tight Junction Assembly. Nat. Cell Biol. 2003, 5, 137–142. [Google Scholar] [CrossRef] [PubMed]
  43. Higashi, T.; Tokuda, S.; Kitajiri, S.; Masuda, S.; Nakamura, H.; Oda, Y.; Furuse, M. Analysis of the “angulin” Proteins LSR, ILDR1 and ILDR2--Tricellulin Recruitment, Epithelial Barrier Function and Implication in Deafness Pathogenesis. J. Cell Sci. 2013, 126, 966–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Tsukita, S.; Yamazaki, Y.; Katsuno, T.; Tamura, A.; Tsukita, S. Tight Junction-Based Epithelial Microenvironment and Cell Proliferation. Oncogene 2008, 27, 6930–6938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Yen, F.T.; Roitel, O.; Bonnard, L.; Notet, V.; Pratte, D.; Stenger, C.; Magueur, E.; Bihain, B.E. Lipolysis Stimulated Lipoprotein Receptor: A Novel Molecular Link between Hyperlipidemia, Weight Gain, and Atherosclerosis in Mice. J. Biol. Chem. 2008, 283, 25650–25659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Stenger, C.; Hanse, M.; Pratte, D.; Mbala, M.L.; Akbar, S.; Koziel, V.; Escanyé, M.C.; Kriem, B.; Malaplate-Armand, C.; Olivier, J.L.; et al. Up-Regulation of Hepatic Lipolysis Stimulated Lipoprotein Receptor by Leptin: A Potential Lever for Controlling Lipid Clearance during the Postprandial Phase. FASEB J. 2010, 24, 4218–4228. [Google Scholar] [CrossRef] [PubMed]
  47. Kohno, T.; Kikuchi, S.; Ninomiya, T.; Kojima, T. The Bicellular Tensile Force Sorts the Localization of LSRs in Bicellular and Tricellular Junctions. Ann. N. Y. Acad. Sci. 2017, 1397, 185–194. [Google Scholar] [CrossRef]
  48. Kohno, T.; Konno, T.; Kojima, T. Role of Tricellular Tight Junction Protein Lipolysis-Stimulated Lipoprotein Receptor (LSR) in Cancer Cells. Int. J. Mol. Sci. 2019, 20, 3555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Martin, T.A.; Jiang, W.G. Loss of Tight Junction Barrier Function and Its Role in Cancer Metastasis. Biochim. Biophys. Acta Biomembr. 2009, 1788, 872–891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Guillot, C.; Lecuit, T. Mechanics of Epithelial Tissue Homeostasis and Morphogenesis. Science (1979) 2013, 340, 1185–1189. [Google Scholar] [CrossRef]
  51. Loza, A.J.; Koride, S.; Schimizzi, G.V.; Li, B.; Sun, S.X.; Longmore, G.D. Cell Density and Actomyosin Contractility Control the Organization of Migrating Collectives within an Epithelium. Mol. Biol. Cell 2016, 27, 3459–3470. [Google Scholar] [CrossRef] [PubMed]
  52. Gumbiner, B.M.; Kim, N.G. The Hippo-YAP Signaling Pathway and Contact Inhibition of Growth. J. Cell Sci. 2014, 127, 709–717. [Google Scholar] [CrossRef] [Green Version]
  53. Mendonsa, A.M.; Na, T.-Y.; Gumbiner, B.M. E-Cadherin in Contact Inhibition and Cancer. Oncogene 2018, 37, 4769–4780. [Google Scholar] [CrossRef] [PubMed]
  54. McClatchey, A.I.; Yap, A.S. Contact Inhibition (of Proliferation) Redux. Curr. Opin. Cell Biol. 2012, 24, 685–694. [Google Scholar] [CrossRef] [PubMed]
  55. Garcia, M.A.; Nelson, W.J.; Chavez, N. Cell–Cell Junctions Organize Structural and Signaling Networks. Cold Spring Harb. Perspect. Biol. 2018, 10, a029181. [Google Scholar] [CrossRef] [Green Version]
  56. Eisenhoffer, G.T.; Rosenblatt, J. Bringing Balance by Force: Live Cell Extrusion Controls Epithelial Cell Numbers. Trends Cell Biol. 2013, 23, 185–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Stramer, B.; Mayor, R. Mechanisms and in Vivo Functions of Contact Inhibition of Locomotion. Nat. Rev. Mol. Cell Biol. 2017, 18, 43–55. [Google Scholar] [CrossRef] [PubMed]
  58. Buckley, C.E.; St Johnston, D. Apical–Basal Polarity and the Control of Epithelial Form and Function. Nat. Rev. Mol. Cell Biol. 2022, 23, 559–577. [Google Scholar] [CrossRef] [PubMed]
  59. Haigis, K.M. KRAS Alleles: The Devil Is in the Detail. Trends Cancer 2017, 3, 686–697. [Google Scholar] [CrossRef]
  60. Li, S.; Balmain, A.; Counter, C.M. A Model for RAS Mutation Patterns in Cancers: Finding the Sweet Spot. Nat. Rev. Cancer 2018, 18, 767–777. [Google Scholar] [CrossRef]
  61. Byron, S.A.; Gartside, M.; Powell, M.A.; Wellens, C.L.; Gao, F.; Mutch, D.G.; Goodfellow, P.J.; Pollock, P.M. FGFR2 Point Mutations in 466 Endometrioid Endometrial Tumors: Relationship with MSI, KRAS, PIK3CA, CTNNB1 Mutations and Clinicopathological Features. PLoS ONE 2012, 7, e30801. [Google Scholar] [CrossRef]
  62. Inoue, S.; Hirota, Y.; Ueno, T.; Fukui, Y.; Yoshida, E.; Hayashi, T.; Kojima, S.; Takeyama, R.; Hashimoto, T.; Kiyono, T.; et al. Uterine Adenomyosis Is an Oligoclonal Disorder Associated with KRAS Mutations. Nat. Commun. 2019, 10, 5785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Satoh, T.; Nishida, M.; Miyazaki, Y.; Sugita, M.; Arai, Y.; Oki, A.; Kono, K.; Tsunoda, H.; Kasahara, K.; Kubo, T. Establishment of a Cisplatin-Resistant New Human Endometrial Adenocarcinoma Cell Line, Sawano Cells. Hum. Cell 1995, 8, 67–72. [Google Scholar] [PubMed]
  64. Ray, I.; Meira, L.B.; Michael, A.; Ellis, P.E. Adipocytokines and Disease Progression in Endometrial Cancer: A Systematic Review. Cancer Metastasis Rev. 2022, 41, 211–242. [Google Scholar] [CrossRef]
  65. Konno, T.; Takano, K.; Kaneko, Y.; Kakuki, T.; Nomura, K.; Yajima, R.; Kakiuchi, A.; Kohno, T.; Himi, T.; Kojima, T. Guanylate Binding Protein-1-Mediated Epithelial Barrier in Human Salivary Gland Duct Epithelium. Exp. Cell Res. 2018, 371, 31–41. [Google Scholar] [CrossRef]
  66. Kyuno, T.; Kyuno, D.; Kohno, T.; Konno, T.; Kikuchi, S.; Arimoto, C.; Yamaguchi, H.; Imamura, M.; Kimura, Y.; Kondoh, M.; et al. Tricellular Tight Junction Protein LSR/Angulin-1 Contributes to the Epithelial Barrier and Malignancy in Human Pancreatic Cancer Cell Line. Histochem. Cell Biol. 2020, 153, 5–16. [Google Scholar] [CrossRef]
  67. Kodera, Y.; Kohno, T.; Konno, T.; Arai, W.; Tsujiwaki, M.; Shindo, Y.; Chiba, H.; Miyakawa, M.; Tanaka, H.; Sakuma, Y.; et al. HMGB1 Enhances Epithelial Permeability via P63/TGF-β Signaling in Lung and Terminal Bronchial Epithelial Cells. Tissue Barriers 2020, 8, 1805997. [Google Scholar] [CrossRef]
  68. Kodera, Y.; Chiba, H.; Konno, T.; Kohno, T.; Takahashi, H.; Kojima, T. HMGB1-Downregulated Angulin-1/LSR Induces Epithelial Barrier Disruption via Claudin-2 and Cellular Metabolism via AMPK in Airway Epithelial Calu-3 Cells. Biochem. Biophys. Res. Commun. 2020, 527, 553–560. [Google Scholar] [CrossRef]
  69. Miyakawa, M.; Konno, T.; Kohno, T.; Kikuchi, S.; Tanaka, H.; Kojima, T. Increase in Epithelial Permeability and Cell Metabolism by High Mobility Group Box 1, Inflammatory Cytokines and TPEN in Caco-2 Cells as a Novel Model of Inflammatory Bowel Disease. Int. J. Mol. Sci. 2020, 21, 8434. [Google Scholar] [CrossRef]
  70. Shindo, Y.; Arai, W.; Konno, T.; Kohno, T.; Kodera, Y.; Chiba, H.; Miyajima, M.; Sakuma, Y.; Watanabe, A.; Kojima, T. Effects of Histone Deacetylase Inhibitors Tricostatin A and Quisinostat on Tight Junction Proteins of Human Lung Adenocarcinoma A549 Cells and Normal Lung Epithelial Cells. Histochem. Cell Biol. 2021, 155, 637–653. [Google Scholar] [CrossRef]
  71. Ohwada, K.; Konno, T.; Kohno, T.; Nakano, M.; Ohkuni, T.; Miyata, R.; Kakuki, T.; Kondoh, M.; Takano, K.; Kojima, T. Effects of HMGB1 on Tricellular Tight Junctions via TGF-β Signaling in Human Nasal Epithelial Cells. Int. J. Mol. Sci. 2021, 22, 8390. [Google Scholar] [CrossRef] [PubMed]
  72. Kojima, T.; Shindo, Y.; Konno, T.; Kodera, Y.; Arai, W.; Miyakawa, M.; Ohwada, K.; Tanaka, H.; Tsujiwaki, M.; Sakuma, Y.; et al. Dysfunction of Epithelial Permeability Barrier Induced by HMGB1 in 2.5D Cultures of Human Epithelial Cells. Tissue Barriers 2022, 10, 1972760. [Google Scholar] [CrossRef]
  73. Nezhat, F.; Datta, M.S.; Hanson, V.; Pejovic, T.; Nezhat, C.; Nezhat, C. The Relationship of Endometriosis and Ovarian Malignancy: A Review. Fertil. Steril. 2008, 90, 1559–1570. [Google Scholar] [CrossRef] [PubMed]
  74. Zhou, W.-J.; Yang, H.-L.; Shao, J.; Mei, J.; Chang, K.-K.; Zhu, R.; Li, M.-Q. Anti-Inflammatory Cytokines in Endometriosis. Cell. Mol. Life Sci. 2019, 76, 2111–2132. [Google Scholar] [CrossRef] [PubMed]
  75. Zeng, Q.; Hong, W. The Emerging Role of the Hippo Pathway in Cell Contact Inhibition, Organ Size Control, and Cancer Development in Mammals. Cancer Cell 2008, 13, 188–192. [Google Scholar] [CrossRef] [Green Version]
  76. Halder, G.; Dupont, S.; Piccolo, S. Transduction of Mechanical and Cytoskeletal Cues by YAP and TAZ. Nat. Rev. Mol. Cell Biol. 2012, 13, 591–600. [Google Scholar] [CrossRef]
  77. Shimada, H.; Abe, S.; Kohno, T.; Satohisa, S.; Konno, T.; Takahashi, S.; Hatakeyama, T.; Arimoto, C.; Kakuki, T.; Kaneko, Y.; et al. Loss of Tricellular Tight Junction Protein LSR Promotes Cell Invasion and Migration via Upregulation of TEAD1/AREG in Human Endometrial Cancer. Sci. Rep. 2017, 7, 37049. [Google Scholar] [CrossRef] [Green Version]
  78. Konno, T.; Kohno, T.; Okada, T.; Shimada, H.; Satohisa, S.; Kikuchi, S.; Saito, T.; Kojima, T. ASPP2 Suppression Promotes Malignancy via LSR and YAP in Human Endometrial Cancer. Histochem. Cell Biol. 2020, 154, 197–213. [Google Scholar] [CrossRef]
  79. Bao, Y.; Nakagawa, K.; Yang, Z.; Ikeda, M.; Withanage, K.; Ishigami-Yuasa, M.; Okuno, Y.; Hata, S.; Nishina, H.; Hata, Y. A Cell-Based Assay to Screen Stimulators of the Hippo Pathway Reveals the Inhibitory Effect of Dobutamine on the YAP-Dependent Gene Transcription. J. Biochem. 2011, 150, 199–208. [Google Scholar] [CrossRef]
  80. Zheng, H.X.; Wu, L.N.; Xiao, H.; Du, Q.; Liang, J.F. Inhibitory Effects of Dobutamine on Human Gastric Adenocarcinoma. World J. Gastroenterol. 2014, 20, 17092–17099. [Google Scholar] [CrossRef]
  81. Paul, S.; Xie, S.; Yao, X.; Dey, A. Transcriptional Regulation of the Hippo Pathway: Current Understanding and Insights from Single-Cell Technologies. Cells 2022, 11, 2225. [Google Scholar] [CrossRef] [PubMed]
  82. Someya, M.; Kojima, T.; Ogawa, M.; Ninomiya, T.; Nomura, K.; Takasawa, A.; Murata, M.; Tanaka, S.; Saito, T.; Sawada, N. Regulation of Tight Junctions by Sex Hormones in Normal Human Endometrial Epithelial Cells and Uterus Cancer Cell Line Sawano. Cell Tissue Res. 2013, 354, 481–494. [Google Scholar] [CrossRef] [PubMed]
  83. Shimada, H.; Satohisa, S.; Kohno, T.; Takahashi, S.; Hatakeyama, T.; Konno, T.; Tsujiwaki, M.; Saito, T.; Kojima, T. The Roles of Tricellular Tight Junction Protein Lipolysis-Stimulated Lipoprotein Receptor in Malignancy of Human Endometrial Cancer Cells. Oncotarget 2016, 7, 27735–27752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Grund, S.; Grümmer, R. Direct Cell-Cell Interactions in the Endometrium and in Endometrial Pathophysiology. Int. J. Mol. Sci. 2018, 19, 2227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Leech, A.O.; Cruz, R.G.B.; Hill, A.D.K.; Hopkins, A.M. Paradigms Lost—An Emerging Role for over-Expression of Tight Junction Adhesion Proteins in Cancer Pathogenesis. Ann. Transl. Med. 2015, 3, 184. [Google Scholar] [CrossRef] [PubMed]
  86. Shimada, H.; Satohisa, S.; Kohno, T.; Konno, T.; Takano, K.; Takahashi, S.; Hatakeyama, T.; Arimoto, C.; Saito, T.; Kojima, T. Downregulation of Lipolysis-Stimulated Lipoprotein Receptor Promotes Cell Invasion via Claudin-1-Mediated Matrix Metalloproteinases in Human Endometrial Cancer. Oncol. Lett. 2017, 14, 6776–6782. [Google Scholar] [CrossRef]
  87. Wang, H.B.; Wang, P.Y.; Wang, X.; Wan, Y.L.; Liu, Y.C. Butyrate Enhances Intestinal Epithelial Barrier Function via Up-Regulation of Tight Junction Protein Claudin-1 Transcription. Dig. Dis. Sci. 2012, 57, 3126–3135. [Google Scholar] [CrossRef]
  88. Dufresne, J.; Cyr, D.G. Activation of an SP Binding Site Is Crucial for the Expression of Claudin 1 in Rat Epididymal Principal Cells. Biol. Reprod. 2007, 76, 825–832. [Google Scholar] [CrossRef] [Green Version]
  89. Honda, H.; Pazin, M.J.; Ji, H.; Wernyj, R.P.; Morin, P.J. Crucial Roles of Sp1 and Epigenetic Modifications in the Regulation of the Cldn4 Promoter in Ovarian Cancer Cells. J. Biol. Chem. 2006, 281, 21433–21444. [Google Scholar] [CrossRef] [Green Version]
  90. Luk, J.M.; Tong, M.K.; Mok, B.W.; Tam, P.C.; Yeung, W.S.B.; Lee, K.F. Sp1 Site Is Crucial for the Mouse Claudin-19 Gene Expression in the Kidney Cells. FEBS Lett. 2004, 578, 251–256. [Google Scholar] [CrossRef]
  91. Oku, N.; Sasabe, E.; Ueta, E.; Yamamoto, T.; Osaki, T. Tight Junction Protein Claudin-1 Enhances the Invasive Activity of Oral Squamous Cell Carcinoma Cells by Promoting Cleavage of Laminin-5 Γ2 Chain via Matrix Metalloproteinase (MMP)-2 and Membrane-Type MMP-1. Cancer Res. 2006, 66, 5251–5257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. van Itallie, C.M.; Fanning, A.S.; Anderson, J.M.; Boireau, S.; Samuel, M.S.; Pannequin, J.; Ryan, J.L.; Choquet, A.; Chapuis, H.; Rebillard, X.; et al. Claudin-1 Regulates Cellular Transformation and Metastatic Behavior in Colon Cancer. J. Clin. Investig. 2005, 115, 1765–1776. [Google Scholar] [CrossRef] [Green Version]
  93. Castro-Castro, A.; Marchesin, V.; Monteiro, P.; Lodillinsky, C.; Rossé, C.; Chavrier, P. Cellular and Molecular Mechanisms of MT1-MMP-Dependent Cancer Cell Invasion. Annu. Rev. Cell Dev. Biol. 2016, 32, 555–576. [Google Scholar] [CrossRef]
  94. Brown, G.T.; Murray, G.I. Current Mechanistic Insights into the Roles of Matrix Metalloproteinases in Tumour Invasion and Metastasis. J. Pathol. 2015, 237, 273–281. [Google Scholar] [CrossRef] [PubMed]
  95. Gaide Chevronnay, H.P.; Selvais, C.; Emonard, H.; Galant, C.; Marbaix, E.; Henriet, P. Regulation of Matrix Metalloproteinases Activity Studied in Human Endometrium as a Paradigm of Cyclic Tissue Breakdown and Regeneration. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2012, 1824, 146–156. [Google Scholar] [CrossRef] [PubMed]
  96. Pandit, A.; das Mahapatra, P.; Saha, P.; Srivastava, A.K.; Swarnakar, S. Interleukin-1β Activated C-FOS Transcription Factor Binds Preferentially to a Specific Allele of the Matrix Metalloproteinase-13 Promoter and Increases Susceptibility to Endometriosis. J. Cell Physiol. 2022, 237, 3095–3108. [Google Scholar] [CrossRef]
  97. Mwaura, A.N.; Riaz, M.A.; Maoga, J.B.; Mecha, E.; Omwandho, C.O.A.; Scheiner-Bobis, G.; Meinhold-Heerlein, I.; Konrad, L. Role of Betaglycan in TGF-β Signaling and Wound Healing in Human Endometriotic Epithelial Cells and in Endometriosis. Biology 2022, 11, 513. [Google Scholar] [CrossRef]
  98. Paz, H.; Pathak, N.; Yang, J. Invading One Step at a Time: The Role of Invadopodia in Tumor Metastasis. Oncogene 2014, 33, 4193–4202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Chang, J.W.; SEO, S.T.; IM, M.A.; WON, H.-R.; LIU, L.; OH, C.; JIN, Y.L.; PIAO, Y.; KIM, H.J.; KIM, J.T.; et al. Claudin-1 Mediates Progression by Regulating EMT through AMPK/TGF-β Signaling in Head and Neck Squamous Cell Carcinoma. Transl. Res. 2022, 247, 58–78. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, K.; Pascal, L.E.; Li, F.; Chen, W.; Dhir, R.; Balasubramani, G.K.; DeFranco, D.B.; Yoshimura, N.; He, D.; Wang, Z. Tight Junction Protein Claudin-1 Is Downregulated by TGF-β1 via MEK Signaling in Benign Prostatic Epithelial Cells. Prostate 2020, 80, 1203–1215. [Google Scholar] [CrossRef] [PubMed]
  101. Stamatovic, S.M.; Johnson, A.M.; Sladojevic, N.; Keep, R.F.; Andjelkovic, A.V. Endocytosis of Tight Junction Proteins and the Regulation of Degradation and Recycling. Ann. N. Y. Acad. Sci. 2017, 1397, 54–65. [Google Scholar] [CrossRef]
  102. Bruewer, M.; Utech, M.; Ivanov, A.I.; Hopkins, A.M.; Parkos, C.A.; Nusrat, A. Interferon-γ Induces Internalization of Epithelial Tight Junction Proteins via a Macropinocytosis-like Process. FASEB J. 2005, 19, 923–933. [Google Scholar] [CrossRef] [PubMed]
  103. Hopkins, A.M.; Walsh, S.V.; Verkade, P.; Boquet, P.; Nusrat, A. Constitutive Activation of Rho Proteins by CNF-1 Influences Tight Junction Structure and Epithelial Barrier Function. J. Cell Sci. 2003, 116, 725–742. [Google Scholar] [CrossRef] [Green Version]
  104. Shen, L.; Turner, J.R. Actin Depolymerization Disrupts Tight Junctions via Caveolae-Mediated Endocytosis. Mol. Biol. Cell 2005, 16, 3919–3936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Matsuda, M.; Kubo, A.; Furuse, M.; Tsukita, S. A Peculiar Internalization of Claudins, Tight Junction-Specific Adhesion Molecules, during the Intercellular Movement of Epithelial Cells. J. Cell Sci. 2004, 117, 1247–1257. [Google Scholar] [CrossRef] [Green Version]
  106. Yamaki, T.; Ohtake, K.; Ichikawa, K.; Uchida, M.; Uchida, H.; Ohshima, S.; Juni, K.; Kobayashi, J.; Morimoto, Y.; Natsume, H. Poly-l-Arginine-Induced Internalization of Tight Junction Proteins Increases the Paracellular Permeability of the Caco-2 Cell Monolayer to Hydrophilic Macromolecules. Biol. Pharm. Bull. 2013, 36, 432–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Murakami, T.; Felinski, E.A.; Antonetti, D.A. Occludin Phosphorylation and Ubiquitination Regulate Tight Junction Trafficking and Vascular Endothelial Growth Factor-Induced Permeability. J. Biol. Chem. 2009, 284, 21036–21046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Krug, S.M.; Hayaishi, T.; Iguchi, D.; Watari, A.; Takahashi, A.; Fromm, M.; Nagahama, M.; Takeda, H.; Okada, Y.; Sawasaki, T.; et al. Angubindin-1, a Novel Paracellular Absorption Enhancer Acting at the Tricellular Tight Junction. J. Control. Release 2017, 260, 1–11. [Google Scholar] [CrossRef] [PubMed]
  109. Pernet-Gallay, K.; Jouneau, P.H.; Bertrand, A.; Delaroche, J.; Farion, R.; Rémy, C.; Barbier, E.L. Vascular Permeability in the RG2 Glioma Model Can Be Mediated by Macropinocytosis and Be Independent of the Opening of the Tight Junction. J. Cereb. Blood Flow Metab. 2017, 37, 1264–1275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Nagahama, M.; Takehara, M.; Kobayashi, K. Interaction of Clostridium Perfringens Iota Toxin and Lipolysis-Stimulated Lipoprotein Receptor (LSR). Toxins 2018, 10, 405. [Google Scholar] [CrossRef] [PubMed]
  111. Sohet, F.; Lin, C.; Munji, R.N.; Lee, S.Y.; Ruderisch, N.; Soung, A.; Arnold, T.D.; Derugin, N.; Vexler, Z.S.; Yen, F.T.; et al. LSR/Angulin-1 Is a Tricellular Tight Junction Protein Involved in Blood-Brain Barrier Formation. J. Cell Biol. 2015, 208, 703–711. [Google Scholar] [CrossRef] [Green Version]
  112. Zeniya, S.; Kuwahara, H.; Daizo, K.; Watari, A.; Kondoh, M.; Yoshida-Tanaka, K.; Kaburagi, H.; Asada, K.; Nagata, T.; Nagahama, M.; et al. Angubindin-1 Opens the Blood–Brain Barrier in Vivo for Delivery of Antisense Oligonucleotide to the Central Nervous System. J. Control. Release 2018, 283, 126–134. [Google Scholar] [CrossRef] [PubMed]
  113. Konno, T.; Kohno, T.; Kikuchi, S.; Shimada, H.; Satohisa, S.; Saito, T.; Kondoh, M.; Kojima, T. Epithelial Barrier Dysfunction and Cell Migration Induction via JNK/Cofilin/Actin by Angubindin-1. Tissue Barriers 2020, 8, 1695475. [Google Scholar] [CrossRef] [PubMed]
  114. Huang, C.; Jacobson, K.; Schaller, M.D. MAP Kinases and Cell Migration. J. Cell Sci. 2004, 117, 4619–4628. [Google Scholar] [CrossRef] [Green Version]
  115. Magi, S.; Tashiro, E.; Imoto, M. A Chemical Genomic Study Identifying Diversity in Cell Migration Signaling in Cancer Cells. Sci. Rep. 2012, 2, 823. [Google Scholar] [CrossRef] [Green Version]
  116. Ichimizu, S.; Watanabe, H.; Maeda, H.; Hamasaki, K.; Ikegami, K.; Chuang, V.T.G.; Kinoshita, R.; Nishida, K.; Shimizu, T.; Ishima, Y.; et al. Cell-Penetrating Mechanism of Intracellular Targeting Albumin: Contribution of Macropinocytosis Induction and Endosomal Escape. J. Control. Release 2019, 304, 156–163. [Google Scholar] [CrossRef] [PubMed]
  117. Bretou, M.; Sáez, P.J.; Sanséau, D.; Maurin, M.; Lankar, D.; Chabaud, M.; Spampanato, C.; Malbec, O.; Barbier, L.; Muallem, S.; et al. Lysosome Signaling Controls the Migration of Dendritic Cells. Sci. Immunol. 2017, 2, eaak9573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Vicente-Manzanares, M.; Ma, X.; Adelstein, R.S.; Horwitz, A.R. Non-Muscle Myosin II Takes Centre Stage in Cell Adhesion and Migration. Nat. Rev. Mol. Cell Biol. 2009, 10, 778–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Masereel, B. An Overview of Inhibitors of Na+/H+ Exchanger. Eur. J. Med. Chem. 2003, 38, 547–554. [Google Scholar] [CrossRef]
  120. Fliegel, L. Structural and Functional Changes in the Na+ /H+ Exchanger Isoform 1, Induced by Erk1/2 Phosphorylation. Int. J. Mol. Sci. 2019, 20, 2378. [Google Scholar] [CrossRef] [PubMed]
  121. Donowitz, M.; Ming Tse, C.; Fuster, D. SLC9/NHE Gene Family, a Plasma Membrane and Organellar Family of Na+/H+ Exchangers. Mol. Aspects Med. 2013, 34, 236–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Koivusalo, M.; Welch, C.; Hayashi, H.; Scott, C.C.; Kim, M.; Alexander, T.; Touret, N.; Hahn, K.M.; Grinstein, S. Amiloride Inhibits Macropinocytosis by Lowering Submembranous PH and Preventing Rac1 and Cdc42 Signaling. J. Cell Biol. 2010, 188, 547–563. [Google Scholar] [CrossRef] [Green Version]
  123. Rotty, J.D.; Wu, C.; Bear, J.E. New Insights into the Regulation and Cellular Functions of the ARP2/3 Complex. Nat. Rev. Mol. Cell Biol. 2013, 14, 7–12. [Google Scholar] [CrossRef] [PubMed]
  124. Mehedi, M.; McCarty, T.; Martin, S.E.; le Nouën, C.; Buehler, E.; Chen, Y.-C.; Smelkinson, M.; Ganesan, S.; Fischer, E.R.; Brock, L.G.; et al. Actin-Related Protein 2 (ARP2) and Virus-Induced Filopodia Facilitate Human Respiratory Syncytial Virus Spread. PLoS Pathog. 2016, 12, e1006062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Denker, S.P.; Huang, D.C.; Orlowski, J.; Furthmayr, H.; Barber, D.L. Direct Binding of the Na-H Exchanger NHE1 to ERM Proteins Regulates the Cortical Cytoskeleton and Cell Shape Independently of H+ Translocation. Mol. Cell 2000, 6, 1425–1436. [Google Scholar] [CrossRef]
  126. Tominaga, T.; Ishizaki, T.; Narumiya, S.; Barber, D.L. P160ROCK Mediates RhoA Activation of Na-H Exchange. EMBO J. 1998, 17, 4712–4722. [Google Scholar] [CrossRef] [Green Version]
  127. Hendus-Altenburger, R.; Pedraz-Cuesta, E.; Olesen, C.W.; Papaleo, E.; Schnell, J.A.; Hopper, J.T.S.; Robinson, C.V.; Pedersen, S.F.; Kragelund, B.B. The Human Na+/H+ Exchanger 1 Is a Membrane Scaffold Protein for Extracellular Signal-Regulated Kinase 2. BMC Biol. 2016, 14, 31. [Google Scholar] [CrossRef] [Green Version]
  128. Amith, S.R.; Wilkinson, J.M.; Baksh, S.; Fliegel, L. The Na+/H+ Exchanger (NHE1) as a Novel Co-Adjuvant Target in Paclitaxel Therapy of Triple-Negative Breast Cancer Cells. Oncotarget 2015, 6, 1262–1275. [Google Scholar] [CrossRef] [Green Version]
  129. Kaminota, T.; Yano, H.; Shiota, K.; Nomura, N.; Yaguchi, H.; Kirino, Y.; Ohara, K.; Tetsumura, I.; Sanada, T.; Ugumori, T.; et al. Elevated Na+/H+ Exchanger-1 Expression Enhances the Metastatic Collective Migration of Head and Neck Squamous Cell Carcinoma Cells. Biochem. Biophys. Res. Commun. 2017, 486, 101–107. [Google Scholar] [CrossRef]
  130. Guan, X.; Luo, L.; Begum, G.; Kohanbash, G.; Song, Q.; Rao, A.; Amankulor, N.; Sun, B.; Sun, D.; Jia, W. Elevated Na/H Exchanger 1 (SLC9A1) Emerges as a Marker for Tumorigenesis and Prognosis in Gliomas. J. Exp. Clin. Cancer Res. 2018, 37, 255. [Google Scholar] [CrossRef]
  131. Yamada, T.; Yoshida, T.; Kawamoto, A.; Mitsuoka, K.; Iwasaki, K.; Tsuge, H. Cryo-EM Structures Reveal Translocational Unfolding in the Clostridial Binary Iota Toxin Complex. Nat. Struct. Mol. Biol. 2020, 27, 288–296. [Google Scholar] [CrossRef] [PubMed]
  132. Amith, S.R.; Fong, S.; Baksh, S.; Fliegel, L. Na+/H+ Exchange in the Tumour Microenvironment: Does NHE1 Drive Breast Cancer Carcinogenesis? Int. J. Dev. Biol. 2015, 59, 367–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Levine, D.A. Integrated Genomic Characterization of Endometrial Carcinoma. Nature 2013, 497, 67–73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Torres-Flores, J.M.; Arias, C.F. Tight Junctions Go Viral! Viruses 2015, 7, 5145–5154. [Google Scholar] [CrossRef]
  135. Commisso, C.; Davidson, S.M.; Soydaner-Azeloglu, R.G.; Parker, S.J.; Kamphorst, J.J.; Hackett, S.; Grabocka, E.; Nofal, M.; Drebin, J.A.; Thompson, C.B.; et al. Macropinocytosis of Protein Is an Amino Acid Supply Route in Ras-Transformed Cells. Nature 2013, 497, 633–637. [Google Scholar] [CrossRef] [Green Version]
  136. Davidson, S.M.; Jonas, O.; Keibler, M.A.; Hou, H.W.; Luengo, A.; Mayers, J.R.; Wyckoff, J.; del Rosario, A.M.; Whitman, M.; Chin, C.R.; et al. Direct Evidence for Cancer-Cell-Autonomous Extracellular Protein Catabolism in Pancreatic Tumors. Nat. Med. 2017, 23, 235–241. [Google Scholar] [CrossRef]
Figure 1. Overview of the endocytic pathways. Phagocytosis incorporates extracellular components through phagocytic receptors. Cells that phagocytose with high efficiency are termed professional cells, whereas those with low efficiency are termed non-professional cells. Pinocytosis nonspecifically incorporates soluble components, such as proteins and growth factors. Small vesicles are formed through clathrin- and caveolae-mediated pathways during pinocytosis. The pathway of extensive incorporation, with membrane protrusion from the cell surface, is termed macropinocytosis. The endometrial cancer cell Sawano exhibits macropinocytosis caused by a transient split of intercellular spaces.
Figure 1. Overview of the endocytic pathways. Phagocytosis incorporates extracellular components through phagocytic receptors. Cells that phagocytose with high efficiency are termed professional cells, whereas those with low efficiency are termed non-professional cells. Pinocytosis nonspecifically incorporates soluble components, such as proteins and growth factors. Small vesicles are formed through clathrin- and caveolae-mediated pathways during pinocytosis. The pathway of extensive incorporation, with membrane protrusion from the cell surface, is termed macropinocytosis. The endometrial cancer cell Sawano exhibits macropinocytosis caused by a transient split of intercellular spaces.
Cancers 14 05056 g001
Figure 2. Schematic diagram of the LSR ligand-induced transient macropinocytosis derived from intercellular split. (Upper panels) In mature epithelial cells, LSR accumulates in apical tricellular junctions. Administration of the LSR ligand splits the adjacent intercellular spaces, leading to macropinocytosis. The intercellular adhesion apparatus was not disrupted by macropinocytosis. G, intercellular gap; TJ, tight junctions; AJ, adherens junctions; D, desmosomes; N, nucleus; Mt, mitochondria. Bar, 1 μm. (Lower panels) The gap between adjacent cells expanded and filled with extracellular fluid, one hour after administration of the LSR ligand. Two hours after addition, numerous macropinosomes that incorporated the extracellular fluid appeared in the cytosol. The vesicles disappeared three hours after addition. The cells then initiated malignant cell growth, resulting in multilayered cell growth. This pathway was suppressed in the presence of a macropinocytosis inhibitor.
Figure 2. Schematic diagram of the LSR ligand-induced transient macropinocytosis derived from intercellular split. (Upper panels) In mature epithelial cells, LSR accumulates in apical tricellular junctions. Administration of the LSR ligand splits the adjacent intercellular spaces, leading to macropinocytosis. The intercellular adhesion apparatus was not disrupted by macropinocytosis. G, intercellular gap; TJ, tight junctions; AJ, adherens junctions; D, desmosomes; N, nucleus; Mt, mitochondria. Bar, 1 μm. (Lower panels) The gap between adjacent cells expanded and filled with extracellular fluid, one hour after administration of the LSR ligand. Two hours after addition, numerous macropinosomes that incorporated the extracellular fluid appeared in the cytosol. The vesicles disappeared three hours after addition. The cells then initiated malignant cell growth, resulting in multilayered cell growth. This pathway was suppressed in the presence of a macropinocytosis inhibitor.
Cancers 14 05056 g002
Figure 3. Schematic representation of the mechanism by which endometrial cancer Sawano initiates malignant cell growth. Mature epithelial sheets, where LSR localize at tricellular tight junctions, suppress macropinocytosis. LSR ligand administration leads to the translocation of LSR from tricellular tight junctions. LSR interacts with Rab5 in the bicellular region, resulting in decreased LSR expression. SLC9A1 is presumably activated through a signal that accompanies the alteration of LSR localization, followed by Rac activation. SLC9A1 scaffolds Raf and ERK downstream of KRAS. Activated Rac induces JNK activation, resulting in the formation of macropinocytosis, followed by an increase in planar cell migration, and eventually, multilayered cell growth.
Figure 3. Schematic representation of the mechanism by which endometrial cancer Sawano initiates malignant cell growth. Mature epithelial sheets, where LSR localize at tricellular tight junctions, suppress macropinocytosis. LSR ligand administration leads to the translocation of LSR from tricellular tight junctions. LSR interacts with Rab5 in the bicellular region, resulting in decreased LSR expression. SLC9A1 is presumably activated through a signal that accompanies the alteration of LSR localization, followed by Rac activation. SLC9A1 scaffolds Raf and ERK downstream of KRAS. Activated Rac induces JNK activation, resulting in the formation of macropinocytosis, followed by an increase in planar cell migration, and eventually, multilayered cell growth.
Cancers 14 05056 g003
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kohno, T.; Kojima, T. Atypical Macropinocytosis Contributes to Malignant Progression: A Review of Recent Evidence in Endometrioid Endometrial Cancer Cells. Cancers 2022, 14, 5056. https://doi.org/10.3390/cancers14205056

AMA Style

Kohno T, Kojima T. Atypical Macropinocytosis Contributes to Malignant Progression: A Review of Recent Evidence in Endometrioid Endometrial Cancer Cells. Cancers. 2022; 14(20):5056. https://doi.org/10.3390/cancers14205056

Chicago/Turabian Style

Kohno, Takayuki, and Takashi Kojima. 2022. "Atypical Macropinocytosis Contributes to Malignant Progression: A Review of Recent Evidence in Endometrioid Endometrial Cancer Cells" Cancers 14, no. 20: 5056. https://doi.org/10.3390/cancers14205056

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