Next Article in Journal
New Insights for an Advanced Understanding of the Molecular Mechanisms in Oral Squamous Cell Carcinoma
Previous Article in Journal
The Total Denervation of the Ischemic Kidney Induces Differential Responses in Sodium Transporters’ Expression in the Contralateral Kidney in Goldblatt Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cellular Regulation of Macropinocytosis

School of Medicine, Jiangsu University, Zhenjiang 212013, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(13), 6963; https://doi.org/10.3390/ijms25136963
Submission received: 16 May 2024 / Revised: 17 June 2024 / Accepted: 23 June 2024 / Published: 26 June 2024
(This article belongs to the Section Molecular Biology)

Abstract

Interest in macropinocytosis has risen in recent years owing to its function in tumorigenesis, immune reaction, and viral infection. Cancer cells utilize macropinocytosis to acquire nutrients to support their uncontrolled proliferation and energy consumption. Macropinocytosis, a highly dynamic endocytic and vesicular process, is regulated by a series of cellular signaling pathways. The activation of small GTPases in conjunction with phosphoinositide signaling pivotally regulates the process of macropinocytosis. In this review, we summarize important findings about the regulation of macropinocytosis and provide information to increase our understanding of the regulatory mechanism underlying it.

1. Introduction

Macropinocytosis is a type of non-selective endocytosis in which extracellular fluid is taken into the cell through the internalization of the plasma membrane (Figure 1) [1,2]. Macropinocytosis is characterized by initiating plasma membrane ruffles on the cell surface where it takes place and subsequently engulfing the extracellular fluid by the formation of giant (0.2–5 µm in diameter) and irregularly shaped endocytic vacuoles, named macropinosomes [3]. The extracellular fluid taken by macropinosomes contains proteins, necrotic cell debris, cell surface receptors, or/and extracellular vesicles (EVs) and provides sources of nutrition for the cell [4]. The contents of macropinosomes are generally transported to lysosomes for degradation.
It has been shown that growth factors, oxidative stress, and starvation are able to trigger macropinocytosis (Figure 1) [4]. Cancer cells under nutritional stress utilize macropinocytosis to obtain nutrients for proliferation and metabolism [5]. Particularly, cancer cells expressing oncogenic RAS mutants (e.g., K-RAS and H-RAS) exhibit high macropinocytosis activity and may develop into methuosis, a type of cell death linked to macropinocytosis [6]. Dendritic cells (DCs) and macrophages utilize macropinocytosis to acquire and present antigens [7]. Viruses and bacteria sometimes invade cells or facilitate uptake by host cells through macropinocytosis [8].
As shown in Figure 1, the general processes of macropinocytosis include four steps: (a) the initiation and activation of macropinocytosis; (b) formation of plasma membrane ruffles and macropinocytic cups; (c) maturation of macropinosomes; and (d) fusion of macropinosomes with lysosomes for the degradation or recycling of macropinosomes. The signaling for the regulation of macropinocytosis normally starts with the activation of RAS protein by macropinocytic stimulators, which in turn initiate signaling cascades involving RAC1 and phosphatidylinositol kinases and their downstream effectors or product that lead to actin cytoskeleton remodeling for plasma membrane ruffle and macropinocytic cup formation [1,9,10,11]. The macropinocytic cups wrap the extracellular fluid and form macropinosomes [1,9]. Macropinosomes are further matured by regulation of the small GTPases RAC1 and CDC42 as well as phosphatidylinositol lipids, and matured macropinosomes undergo recycling or degradation processes.
In this review, we focus on the regulatory process of macropinocytosis and hopefully increase our understanding of the regulatory mechanism underlying the macropinocytic process.

2. Signaling Pathways for Initiation and Activation of Macropinocytosis

The small GTPase RAC1 was the first protein found to initiate plasma membrane ruffles and macropinocytosis [12]. Subsequently, numerous studies connected plasma membrane ruffles to the initiation of macropinocytosis and identified RAC1 as a key regulatory protein in the initiation of macropinocytosis and the formation of macropinosomes. The pro-oncogenic protein RAS, a small GTPase upstream of RAC1 and PI-3 kinase, has been identified as an essential regulatory protein for macropinocytosis in the nutrition acquisition of cancer cells [13]. The RAS/RAC/PI3K signaling axis is currently considered as the pivotal regulatory pathway for generating plasma membrane ruffles, initiating macropinocytosis, and forming macropinosomes.
It has been observed that amino acid starvation activates macropinocytosis in cancer cells through the AMPK, YAP, or EGFR signaling pathways [14,15,16]. It seems that all these signaling pathways point to the activation of RAS and/or RAC. In addition to EGFR, multiple tyrosine kinase receptors, such as hepatic growth factor receptor (c-Met), platelet-derived growth factor receptor (PDGFR), colony-stimulating factor-1 receptor (CSF-1R), and AXL, also activate macropinocytosis [17,18,19,20]. Macropinocytosis is also activated by Wnt signaling [21,22] and immune stimulating signaling, such as interleukin signaling [23]. These signaling pathways activate downstream K-RAS or H-RAS and RAC, leading to the remodeling of the actin cytoskeleton and the formation of membrane ruffles. Macropinocytosis is intensely reliant on the phosphoinositide 3-kinase (PI3K) isoform PI3Kβ [24]. It has been demonstrated that macropinocytosis is impaired by PI3K inhibitors, such as GDC-0941, ZSTK474, wortmannin, and LY294002 [10,20,25].
In immune cells, such as DCs and macrophages, macropinocytosis may constitutively takes place [23,26]. In comparison, macropinosomes from constitutive macropinocytosis, which is dependent on calcium-sensitive receptors (CaSRs), tend to be smaller than those induced by growth factors [27]. CaSR, a G protein-coupled receptor (GPCR) [28], responds to extracellular calcium and initiates a signaling cascade that promotes the constitutive expansion of membrane ruffles and the formation of macropinosomes [27].

3. Regulation of Macropinosomal Formation

The initiation of macropinocytosis commences with the formation of membrane ruffles. There are two types of plasma membrane ruffles: peripheral ruffles (PRs) and circular dorsal ruffles (CDRs) [29,30], both of which are able to develop into macropinosomes. During the process of forming plasma membrane ruffles, the remodeling of the actin cytoskeleton is a critical step, as evidenced by the fact that actin polymerization disruptive reagents (e.g., cytochalasin D) inhibit the formation of macropinosomes [31]. The RHO small GTPase family member RAC plays the most important role in the remodeling of the cytoskeleton for membrane ruffles. The plasma membrane inositol phospholipids are key players in actin cytoskeleton remodeling, activating RAC and recruiting actin cytoskeletal regulatory proteins. Phosphatidylinositol-3,4,5-trisphosphate (PI(3,4,5)P3) is able to recruit the guanine nucleotide exchange factors T-lymphoma invasion and metastasis 1 (TIAM1) and dedicator of cytokinesis 2 (DOCK2) to the plasma membrane which subsequently activates RAC [32]. Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), which activates the suppressor of cAMP receptor (SCAR)/WASP family verprolin homologous (WAVE) complex, and the RAC effector Wiskott–Aldrich syndrome protein (WASP), which interacts with PI(4,5)P2, actin, and actin-related proteins 2/3(Arp2/3), coordinate the assembly of the Arp2/3 complex that promotes actin polymerization [28], leading to the rearrangement of the actin cytoskeleton for the formation of ruffles [2].
The formation of CDRs is different from that of PRs, as it requires extra molecular machines, such as RAB5 GTPase, for the formation of circular ruffles in addition to RAS and RAC [30]. It was observed that the constitutively active mutant of RAS or RAC1 alone was not enough to induce CDRs [2]. The overexpression of the inactive mutant of RAB5, RAB5-S34N, inhibits circular ruffle formation in PDGF-stimulated mouse embryonic fibroblast (MEF) cells [33]. Increasing the expression of RAB5 together with the active form of RAS enhances the formation of circular ruffles [33,34]. Upon the formation of circular ruffles, they are closed by ruffle-like motility, sequestering large amounts of extracellular fluid in macropinocytic cups, and form macropinosomes when these ruffles fuse with the plasma membrane and encapsulate large amounts of extracellular fluid.
Multiple small GTPases, including RHO family GTPases, ARF GTPases, RAB GTPases, and inositol phospholipids, play pivotal roles in the process of macropinocytic cup closure and macropinosomal formation. RAC1 is one of the most important small GTPases for the regulation of membrane ruffles and macropinosomal formation in a wide range of cell types, including DCs, macrophages, fibroblasts, and epithelial cells [10]. RAC1 activates actin nucleation by interacting with downstream effectors such as PAK1 and WASP for the remodeling of the actin cytoskeleton and promotes the formation of ruffles and macropinocytic cups in the plasma membrane [28,35]. RAC1 is inactivated when macropinocytic cups are occluded to form macropinosomes [36]. RHOG is homologous to RAC1 and functions as an upstream activator of RAC1 [37,38]. It was observed that the RHOG-mediated activation of RAC1 is crucial for the formation of the macropinocytic cup in fibroblasts and A431 cells [10,37,39]. RHOC is transiently activated at the circular ruffles prior to the closure of macropinocytic cups for the formation of macropinosomes [40]. RHOA activation was observed following the closure of macropinocytic cups [41]. The ARF family members ARF6 and ARF1 also play important roles in macropinocytosis. Upon RAC1 activation, ARF6 elicits the accumulation of ARNO (ARF-guanine nucleotide exchange factor GEF) for the activation of ARF1 and actin cytoskeletal remodeling [42]. Furthermore, ARF6 activates phospholipase D1, which is required for macropinosomal formation [10]. During macropinosomal formation, RAB5a is activated and recruited to the macropinocytic site. By interacting with its downstream effector, Rabankyrin-5, RAB5 regulates actin cytoskeletal remodeling and promotes membrane ruffles and macropinosomal formation [42]. There are studies claiming that RAB34 engages in membrane ruffling and regulates the formation of macropinosomes [43,44], but this is generally contingent on the activity of other small GTPases, such as RAS [45] and RAC1 [43].
Inositol phospholipids, including PI(4,5)P2, PI(3,4,5)P3, and phosphatidylinositol 3,4-bisphosphate (PI(3,4)P2), are involved in macropinosomal formation [42]. A marked increase in PI(4,5)P2 levels has been reported when plasma membrane ruffling occurs [46]. PI(4,5)P2 recruits various actin-binding proteins to the plasma membrane and initiates actin polymerization and reorganization, which are necessary for membrane ruffling. The small GTPase RAC1 or ARF6 promotes the production of PI(4,5)P2 by activating PI4P5K, the enzyme that phosphorylates phosphatidylinositol 4-phosphate (PI4P) to form PI(4,5)P2 [47]. Upon membrane ruffle closure to form macropinocytic cups, the level of PI(4,5)P2 at the ruffles decreases along with a significant increase in the level of PI(3,4,5)P3 [3,48]. PI(3,4,5)P3 binds to the PH domain of multiple actin remodeling proteins that activate their downstream signaling effectors for actin cytoskeletal reconstruction [10]. Subsequent to macropinocytic cup closure, PI(3,4,5)P3 is dephosphorylated to form PI(3,4)P2 by SHIP 5-phosphatase [46].
Different from other forms of endocytosed vesicles, macropinosomes have no apparent cortical structure and are considerably larger. Since macropinosomes can be as large as 5 μm in diameter, they can easily be identified by using liquid phase markers, such as lucifer yellow (LY), horseradish peroxidase, or dextran [29,49].

4. Regulation of Maturation and Transport of Macropinosomes

Macropinosomes shrink as they mature, allowing the cell to resolve massive amounts of internalized material by macropinocytosis [50]. A loss of fluid in macropinosomes is required to maintain cell size and ionic composition. The contraction of macropinosomes is regulated by the two-pore channel (TPC)-mediated Na+ efflux and the Cl channel, namely ClC-5, together with the pH-regulated ASOR/TMEM206 channel-mediated Cl efflux [50]. TPC is a transmembrane channel dedicated to Na+ transport [50] and induces cell water loss through osmosis [42]. Thus, TPC is capable of reducing the hydrostatic pressure of macropinosomes [42]. TPC is regulated by PI(3,5)P2, an inositol phospholipid product of the phosphoinositide kinase, FYVE-type zinc finger containing (PIKfyve) [51], and mTOR kinase [52,53]. PI(3,5)P2 is a potent activator of TPC and promotes the process of macropinocytosis; thus, PIKfyve kinase activity directly controls macropinocytosis [51]. mTOR phosphorylates TPC when the cell has adequate nutrition and the phosphorylated TPC is inactivated; thus, macropinocytosis is inhibited [52,53]. When the cell is in starvation condition, mTOR is inactive, and the TPC channel is open, the macropinocytotic process is activated. Water loss due to TPC not only causes a decrease in the size of macropinosomes but also induces the formation of tubular extensions of macropinosomes [3]. The tubules detach from the macropinosomes by membrane splitting and are subsequently sorted and transported to specific intracellular compartments in a microtubule-dependent transport mode [54].
RAB GTPases are actively involved in macropinosomal transport [55]. As a molecular switch, RAB GTPases interact with downstream effectors to regulate various aspects during macropinosomal trafficking [56]. RAB5 is known for the regulation of the early stage of macropinocytosis and co-localization with early macropinosomes. It seems that the dissociation of RAB5 from early macropinosomes is a key step for the maturation and transport of macropinosomes. One of the determination processes is the RAB5/RAB7 transition [10]; i.e., RAB5 is replaced with RAB7 on macropinosomes for their maturation and lysosomal transport. It has been shown that active RAB7 interacts with amyotrophic lateral sclerosis 2 (ALS2), an activator for RAB5, dissociates ALS2 from RAB5, and inactivates RAB5 [57], thus replacing RAB5 on macropinosomes [57]. In phagocytic cells, the recycling of macropinosomes is specifically regulated by the RAB10/ leucine-rich repeat kinase (LRRK) signaling axis [58]. The RAB10 effector EH domain binding protein 1-like 1 (EHBP1L1) mediates early macropinosomal recycling and the phosphorylation of RAB10 by LRRK blocking the recycling effect of EHBP1L1. This phosphorylation enables early macropinosomes to avoid recycling back to the plasma membrane and the further maturation of macropinosomes, which is crucial for macropinosome-mediated immune responses [58]. In addition, RAB20 and RAB21 may be involved in the maturation of macropinosomes [59,60]. It was observed that RAB21 and RAB20 were recruited to the membrane of early macropinosomes. While RAB21 dissociated from Lamp1-positive macropinosomes, RAB20 remained on the RAB7 and Lamp1-positive macropinosomes, suggesting that RAB20 may directly participate in the maturation of macropinosomes [59,60]. However, the exact role of RAB20/RAB21 in the maturation of macropinosomes is still not clear.
Nevertheless, as RAB GTPases regulate processes in the transport of various types of membrane vesicles, how to sort out the specific roles of RAB GTPases in macropinosomal maturation and transport is a big challenge for future studies.

5. Regulation of Degradation Process of Macropinocytosis

The matured macropinosomes undergo either recycling or degradation. It has also been proposed that the transport destination of macropinosomes is determined by the context of signaling pathways in macropinocytosis [61]. Macropinosomes derived from RAS- or RAC-activated macropinocytosis are primarily transported to lysosomes, whereas ones from ARF6-activated macropinocytosis are recycled back to the cell surface [62]. The proportion of those undergoing recycling versus lysosomal degradation may vary among cell types, cargos, and stages of macropinocytosis [63].
Similar to endosomal trafficking, macropinosomal trafficking requires vesicle acidification. The disruption of vesicle or lysosomal acidification inhibits macropinocytosis and macropinosomal transport. Hydroxychloroquine (HCQ), a lysosomal inhibitor, inhibits macropinocytosis by interrupting lysosomal acidification [64]. The vesicular H(+)-ATPase (V-H(+)-ATPase), as a proton pump, is essential for maintaining vesicle acidification [65]. The V-H(+)-ATPase inhibitor bafilomycin A1 inhibits the vacuolization of macropinosomes [66] and blocks macropinosome–lysosome fusion during macropinocytosis [67]. It was observed that bafilomycin A1 blocks the formation of plasma membrane ruffling in macropinocytosis, suggesting that V-H(+)-ATPase may reside in the plasma membrane and regulate extracellular pH levels, which affect the formation of plasma membrane ruffles [31]. In fact, V-H(+)-ATPase is localized at the plasma membrane of the K-RAS mutant PDAC cells [68,69]. In addition, the downregulation of PIKfyve prevents the fusion of late endosomes or lysosomes with macropinosomes containing EGFR, and the effect of PIKfyve may be mediated by its downstream effector transient receptor potential mucolipin 1 (TRPML1), which was observed to modulate the size of vacuoles [42,52]. It has been shown that PI(3,5)P2, the product of PIKfyve, regulates the lysosomal Ca2+ channel which is crucial for lysosomal membrane fusion [3]. PI(3,5)P2 mediates macropinosome–lysosome fusion through the activation of TRPML1 [70]. Furthermore, when TRPML1 is inactivated upon the low activity of PIKfyve, the activation of mTORC1 is induced and the catabolism of macropinosomes is restrained [71]. These data indicate that PIKfyve and TRPML1 play important roles in macropinosome–lysosome fusion and the degradation of macropinosomes. In addition, it was observed that septins associate with matured macropinosomes in a PI(3,5)P2-dependent manner and facilitate the transportation and fusion of macropinosomes with lysosomes [50]. The knockdown of septin2 causes the accumulation of macropinosomes and formation of large vacuoles. This study indicates that septin2 is a key regulator in macropinosome maturation and lysosomal degradation. However, the molecular mechanism underlying the promotion of septins of macropinosomal trafficking to and fusion with lysosomes currently remains unknown.

6. Vacuolization of Macropinosomes and Methuosis

Macropinosomes normally undergo a maturation process by which they are either degraded by late endosomal/lysosomal processes or recycled back to the plasma membrane for supplying nutrients and maintaining cell membrane balance [29]. However, when abnormally active macropinocytosis produces excessive macropinosomes that are unable to fuse with lysosomes and recycle back to the plasma membrane, the macropinosomes fuse with each other and form huge vacuoles in the cell that have no markers of autophagosomes (LC3), early endosomes (RAB5 and EEA1), or recycling endosomes (RAB11) [72] but have markers of late endosomes and lysosomes (RAB7 GTPase and membrane glycoprotein Lamp-1). The vacuoles lack hydrolases, are not acidic, and have no late endosomal function [73]. This vacuolization results in cell death, which is called methuosis.
Methuosis was initially observed in the glioblastoma cell overexpression of an oncogenic RAS mutant [74]. Methuosis is a novel form of non-apoptotic cell death characterized by the accumulation of large vacuoles derived from macropinosomes that eventually lead to membrane rupture, cell lysis, and death [70]. This unique cell death with morphological features is distinct from both apoptosis and other non-apoptotic cell death forms, such as autophagy and necrosis [75]. Methuosis usually occurs in tumor cells, such as non-small cell lung cancer cells (A549), prostate cancer cells (DU145), gastric cancer cell lines (HGC-27), and HeLa cells, but sometimes also occurs in normal cells (e.g., primary chicken cardiomyocytes and fibroblasts) with overloaded vesicles [76].
Studies have shown that methuosis has no connection with RAS/MAPK signaling [77,78]. Moreover, the process of methuosis does not depend on the activity of class I PI3K, the RHOA GTPases, or the RALA GTPase but on the downstream effectors of the oncogenic RAS [73]. RAC is the downstream signaling molecule of RAS that drives methuosis [79]. The unrestricted activation of RAC1 by RAS (G12V) stimulates the formation of plasma membrane ruffles and macropinosomes. At the same time, activated RAC1 interacts with GIT1, a GAP protein of ARF6, and causes the inactivation of ARF6 [78]. As ARF6 plays a role in the promotion of the recycling of macropinosomes to the plasma membrane [78], the inactivation of ARF6 by RAC1 reduces the recycling of macropinosomes. Consequently, excessive macropinosomes accumulate within the cell, fuse with each other, and form extremely large vacuoles. These large vacuoles are not functional and degradable, ultimately leading to methuosis [42].
In recent years, several methuosis inducers have been identified. All of these inducers have shown killing effects on cancer cells. This has increased interest in the application of methuosis inducers for cancer therapy. The prototype compounds 3-(5-methoxy-2-methyl-1H-indol-3-yl-3-yl)-1-(4-pyridyl)-2-propen-1-one (abbreviated as MOPPEP) and chalcone 3-(2-methyl-1H indol-3-yl)-1-(4-pyridinyl)-2-propen-1-one (abbreviated as MIPP) were reported to induce methuosis in cancer cell lines at low micromolar concentrations [75,80]. A recent study identified PIKfyve as the specific target protein of MOMIPP in the induction of methuosis [81]. PI(3,5)P2, the product of PIKfyve, is known to play a key role in late-stage macropinosomal trafficking and may mediate the effect of MOMIPP on cellular methuosis [82,83]. However, excessive vacuolization and methuosis induced by inducer chemicals seem to be independent of the RAS-RAC1 signaling pathway [76].

7. Macropinocytosis and Cancer Therapy

Three anticancer strategies based on macropinocytosis have been developed, including delivering chemo/targeted therapeutic drugs to cancer cells via macropinocytosis [9,84,85]; inducing methuosis in cancer cells with the excessive activation of macropinocytosis [86,87,88]; and inhibiting macropinocytosis to block nutrient uptake in cancer cells [89,90,91]. Currently, utilizing macropinocytosis to deliver therapeutic drugs or agents is the major strategy being applied in cancer therapy. Macropinocytosis-based anti-cancer therapy is applied in chemo/targeted therapy, immunotherapy, and nucleic acid therapy either by macropinocytic delivery of anti-cancer chemo/targeted drugs, antibodies, or vaccines or nucleic acids into cancer cells, or by the regulation of macropinocytosis using macropinocytic inhibitors or activators to induce nutritional stress or methuosis [92].
(1) Macropinocytosis-associated chemo/targeted therapy. Macropinocytosis-associated chemo/targeted therapy includes two aspects: one is the utilization of macropinocytosis to deliver anticancer chemo/targeted drugs or agents into cancer cells to produce anti-cancer effects. The other is to employ macropinocytic inhibitors or activators to inhibit or over-activate macropinocytosis to either induce nutritional stress or methuosis of cancer cells. The chemo/targeted drugs or agents that currently used for cancer therapy by either utilizing macropinocytosis for delivering or inhibiting macropinocytosis are shown in Table 1.
RAS mutant cancer cells have very active macropinocytosis [9]. Thus, the utilization of the hyper-active macropinocytosis to deliver chemo drugs into cancer cells is an efficient means for cancer therapy. It has been demonstrated that chemo/targeted drugs can bind to albumin and enter KRAS-mutated PDAC cells via macropinocytosis [85,93]. Paclitaxel (PTX) is a chemotherapeutic drug for multiple types of cancer. To facilitate the uptake of PTX by RAS mutant cancer cells, PTX is formulated with albumin nanoparticles [84]. The albumin-nanoparticle-wrapped PTX is internalized through active macropinocytosis, resulting in the inhibition of cancer cells’ growth [84]. With a similar strategy, other chemo drugs or anti-cancer agents, such as doxorubicin, human β-defensin-2 peptide, and triptolide, were used in cancer therapy [9]. Tubeimoside-1 (TBM1), a natural triterpenoid saponin found in the traditional Chinese herbal medicine Bolbostemmatis Rhizoma, is capable of acting as a drug transporter by stimulating macropinocytosis [86]. Furthermore, the β-defensin-2-loaded human serum albumin (DF-HSA) [94], gefitinib-loaded EVs [95], and triptolide prodrug-loaded UPSM (T-UPSM) [96] are all effectively delivered into cancer cells by macropinocytosis for cancer therapy.
Few specific macropinocytic inhibitors have been identified so far for cancer therapy. 5-(N-ethyl-N-propyl) amiloride (EIPA), the inhibitor of Na+/H+-exchanger 3 (NHE3), is commonly used for inhibiting macropinocytosis [97]. A recent study found that EIPA combined with the proteasomal inhibitor bortezomib packed in pH-responsive polymersomes was able to effectively inhibit the growth of lung cancer A549 cell-xenografted mouse tumors [98]. As shown in Table 1, the vesicular H+-ATPase inhibitor Bafilomycin A1 [66,97], the PAK1 inhibitor IPA-3 [22], the actin polymerization inhibitors [31], the PI3K inhibitors [25], the DOCK1 inhibitor TBOPP [99] and the lysosomal inhibitor HCQ [64] are all able to inhibit macropinocytosis and have been used for cancer therapy. The mTORC1/2 inhibitors OSI-027, PP242 [100], and Torin 1 [101,102], the CK2 inhibitor silmitasertib [87,103], an ursolic-acid-derived small molecule (compound 17) [104], the quinolone derivative Vacquinol-1 [105], Platycodin D (PD) [106], jaspine B (JB) [107], and Epimedokoreanin C (EKC) [108] have been used for anti-cancer therapy via the promotion of macropinosomal vacuolization and by inducing methuosis, thereby killing cancer cells.
(2) Macropinocytosis-associated immunotherapy. Macropinocytosis-associated immunotherapy either delivers anti-cancer antibodies or vaccines into cancer cells by macropinocytosis or uses immune molecules to specifically target macropinocytosis [109]. Examples of macropinocytosis-associated immunotherapy are listed in Table 1. The Fv-LDP-D3 construct, which comprises the scFv antibody and albumin structural domains, has been shown to promote the efficient internalization of anticancer drugs via macropinocytosis [110]. It was observed that the interaction of CD99 with 0662-mAb induces methuosis by over-activating the IGF-1R/RAS/RAC1 signaling cascade [79]. AGS-16C3F, an antibody-drug conjugate (ADC) against ectonucleotide pyrophosphatase/phosphodiesterase 3 (ENPP3), is currently in development for the treatment of metastatic renal cell carcinoma [111]. AGS-16C3F is internalized in cancer cells via macropinocytosis and thus inhibits cancer cell proliferation [111]. The bacillus Calmette–Guerin (BCG) vaccine for tuberculosis (TB) has been shown to be effective in the treatment of bladder cancer by macropinocytic delivery, which is dependent on RAC1, CDC42, and its effector PAK1 [112]. The mycobacterium tuberculosis vaccine (MTBVAC) is now being developed as a new immunotherapeutic drug for bladder cancer [113]. A biomimetic nanovaccine R837-αOVA-ApoE3-HNP was created for cancer immunotherapy [114]. It contains a poly-(D, l-lactide-co-glycolide) (PLGA) core to encapsulate adjuvant imiquimod (R837), a phospholipid membrane to load antigen peptide (αOVA), and apolipoprotein E3 (ApoE3), to facilitate the internalization of cancer antigens into DCs. It has been demonstrated that the R837-αOVA-ApoE3-HNP mimetic nanovaccine is effectively internalized by the macropinocytosis of dendritic cells, resulting in the activation of antigen-specific T cells and the inhibition of tumor metastasis in a mouse model [114].
(3) Macropinocytosis-associated nuclear acid therapy. The anti-cancer agents delivered by macropinocytosis are not limited to chemo drugs or proteins. Anti-cancer RNAs, such as anti-cancer miRNAs and anti-sense RNAs, formulated with exosomes, liposomes, or polymers, can be delivered into cancer cells through macropinocytosis. Macropinocytosis-associated nucleic acid therapy utilizes macropinocytosis to deliver nanoparticle-, lipid-, or polymer-formulated anti-cancer nucleic acids into cancer cells for cancer therapy [115,116] (Table 1). Although nucleic acid therapy has not been developed as much as the other therapies, its potential benefits have been increasingly noted. For instance, exosomes containing specific siRNA or shRNA against oncogenic KRAS-G12D inhibit the growth of pancreatic cancer through macropinocytosis [117]. It has been shown that CXCR4-receptor-stimulated lipoprotein-like nanoparticle-loaded miRNA-34a is internalized by the macropinocytosis of glioma-initiating cells, effectively inhibits their stemness and chemo-resistance, and enhances the survival of mice with glioma-initiating cells [118]. Cancer cells can take up nanostructures carrying activating transcription factor-5 (ATF5) siRNA through macropinocytosis, leading to apoptosis [115]. Furthermore, it was observed that the antisense oligonucleotides of translationally controlled tumor protein (TCTP) with the lipid-modified 5′ end were effectively internalized by the macropinocytosis of castration-resistant prostate cancer cells (CRPCs) [119]. This macropinocytic delivery of TCTP antisense oligonucleotides down-regulated the expression of TCTP, reduced the level of cancer cell viability, and significantly delayed CRPC-xenografted tumor progression in a mouse model [119].
Table 1. Therapeutic modalities exploiting macropinocytosis in cancers.
Table 1. Therapeutic modalities exploiting macropinocytosis in cancers.
Therapeutic
Modalities
DrugsMechanismsCancersReferences
Chemo/targeted
therapy
PTXDrug delivery via macropinocytosisRAS-driven
cancer
[84]
TBM1Drug delivery via macropinocytosisColon cancer[86]
DF-HSADrug delivery via macropinocytosisRAS-driven
cancer
[94]
gefitinibDrug delivery via macropinocytosisLung cancer[95]
T-UPSMDrug delivery via macropinocytosisPDAC[96]
EIPAInhibition of NHE3 to influence submembrane pHRAS-driven
cancer
[97,98]
Bafilomycin A1Impairment of
lysosomal pH
RAS-driven
cancer
[66,97]
IPA-3Inhibition of actin polymerizationRAS-driven and Wnt-driven cancer[22]
cytochalasin DInhibition of actin polymerizationRAS-driven
cancer
[31]
wortmanninInhibition of PI3K signaling pathwayRAS-driven
cancer
[25]
LY294002Inhibition of PI3K signaling pathwayRAS-driven
cancer
[25]
TBOPPPreventing DOCK1 from activating RAC1RAS-driven
cancer
[99]
HCQBlocking lysosomal acidificationPancreatic
cancer
[64]
OSI-027Induction of macropinocytosisBrain cancer, colorectal cancer, cervical cancer, breast cancer, lung cancer[100]
PP242Induction of macropinocytosisBrain cancer,
colorectal cancer, cervical cancer, breast cancer, lung cancer
[100]
Torin 1Induction of macropinocytosisRAS-driven
cancer
[101,102]
silmitasertibInduction of macropinocytosisColorectal
cancer, oral squamous cell
carcinoma
[87,103]
compound 17Induction of macropinocytosisCervical cancer, liver cancer,
fibrosarcoma, breast adenocarcinoma, neuroblastoma
[104]
Vacquinol-1Induction of
methuosis
Glioblastoma[105]
PDInduction of
methuosis
A549 lung cancer cell, MCF7 breast cancer cell[106]
JBInduction of
methuosis
HGC-27
Stomach cancer cell
[107]
EKCInduction of
methuosis
NCI-H292 and A549 lung
cancer cells
[108]
MOMIPPInduction of
methuosis
Glioblastoma[75,80]
ImmunotherapyFv-LDP-D3Targeting EGFRPancreatic
cancer
[110]
CD99 with 0662mAbInduction of
methuosis
Ewing sarcoma[79]
AGS-16C3FUsing
macropinocytosis
Metastatic renal cell carcinoma[111]
BCGUsing
macropinocytosis
Bladder cancer[22,112]
MTBVACUsing
macropinocytosis
Bladder cancer[113]
R837-αOVA-ApoE3-HNP bionic nanovaccineUsing
macropinocytosis
Lung cancer[114]
Nucleic acid therapyKRAS-G12D siRNADownregulation of KRAS-G12D
expression
Pancreatic
cancer
[117]
miRNA-34aUsing
macropinocytosis
Glioma[118]
TCTP ASOsDownregulation of TCTP expressionProstate cancer[119]
ATF5 siRNAUsing
macropinocytosis
Glioblastoma[115]

8. Conclusions

In summary, as shown in Figure 1, macropinocytosis initiates with the formation of plasma membrane ruffles, followed by the production of macropinocytic cups, which in turn generate early macropinosomes. The early macropinosomes continue to mature into late macropinosomes, which fuse with lysosomes and degrade their cargos. Some of the early macropinosomes are able to recycle back to the plasma membrane. Excessive macropinosomes produced by overactive macropinocytosis may fuse with each other to form large vacuoles, leading to methuosis and killing the cell. Therefore, macropinocytosis is a dynamic process with multiple signaling pathways involved. The central signaling molecules involved in the activation of macropinocytosis are RAS, RAC1, and PI-3 kinases. Many signaling pathways, including growth factors, nutrient stress, hypoxia, and pathogen invasion, activate or regulate macropinocytosis through signaling convergence to the RAS, RAC1, and/or PI-3 kinases.
As an important cellular metabolic process, macropinocytosis is associated with other metabolic process, such as autophagy, for cell survival and growth, particularly for cancer cell survival and growth under nutritional or oxidation stress. RAS, the key regulatory protein of macropinocytosis, is a known oncogenic protein with a high tumorigenic mutation frequency in solid tumors. This has increased interest in macropinocytosis regarding its function in cancer. Many studies have shown that macropinocytosis plays an oncogenic role in cancer cells. Thus, targeting or utilizing macropinocytosis is an important strategy to improve current cancer therapeutic outcomes, especially for RAS mutation-mediated tumors.

Author Contributions

Q.L. supervised and participated in the writing and revision of the manuscript. Y.W. collected the references and wrote the manuscript; X.H. and Z.W. participated in the collection of the references and the revision of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work is supported by National Natural Science Foundation of China (grant numbers 82172942 and 81871888 to Q.L.).

Acknowledgments

We would like to thank Wannian Yang for his critical reading of and comments on this manuscript.

Conflicts of Interest

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

Abbreviations

EVs, extracellular vehicles; DCs, dendritic cells; PAK1, P21 Protein Activated Kinase 1; PI, Phosphatidylinositol; EGFR, epidermal growth factor receptor; PDGF, platelet-derived growth factor receptor; CSF-1R, colony-stimulating factor-1 receptor; PI3K, phosphoinositide 3-kinase; CaSR, calcium-sensing receptor; GPCR, G protein-coupled receptor; DAG, diacylglycerol; PLC, phospholipase C; PKC, protein kinase C; SFK, Src-family kinases; PA, phosphatidic acid; PRs, peripheral ruffles; CDRs, circular dorsal ruffles; PI(3,4,5)P3, Phosphatidylinositol-3,4,5-trisphosphate; TIAM1, T-lymphoma invasion and metastasis 1; DOCK2, dedicator of cytokinesis 2; PI(4,5)P2, Phosphatidylinositol 4,5-bisphosphate; SCAR, suppressor of cAMP receptor; WAVE, WASP family verprolin homologous; WASP, Wiskott–Aldrich syndrome protein; Arp2/3, Actin-Related Proteins2/3; MEF, Mouse Embryo Feeder; PI(3,4)P2, phosphatidylinositol 3,4-bisphosphate; PI4P, phosphatidylinositol 4-phosphate; LY, Lucifer yellow; TPC, two-pore channel; PIKfyve, phosphoinositide kinase, FYVE-type zinc finger containing; ALS2, Amyotrophic lateral sclerosis 2; LRRK, leucine-rich repeat kinase; EHBP1L1, EH domain binding protein 1-like 1; HCQ, Hydroxychloroquine; V-H(+)-ATPase, vesicular H(+)-ATPase; TRPML1, Transient receptor potential mucolipin 1; PTX, paclitaxel; TBM1, tubeimoside-1; T-UPSM, Triptolide prodrug-loaded UPSM; EIPA, 5-(N-ethyl-N-propyl) amiloride; NHE3, Na+/H+-exchanger 3; PD, Platycodin D; JB, Jaspine B; EKC, Epimedokoreanin C; ADC, antibody-drug conjugate; ENPP3, ectonucleotide pyrophosphatase/phosphodiesterase 3; BCG, Bacille Calmette-Guérin; TB, tuberculosis; MTBVAC, mycobacterium tuberculosis vaccine; ATF5, activating transcription factor-5; TCTP, translationally controlled tumor protein; CRPCs, castration-resistant prostate cancer cells.

References

  1. Swanson, J.A. Shaping cups into phagosomes and macropinosomes. Nat. Rev. Mol. Cell Biol. 2008, 9, 639–649. [Google Scholar] [CrossRef] [PubMed]
  2. Kerr, M.C.; Teasdale, R.D. Defining macropinocytosis. Traffic 2009, 10, 364–371. [Google Scholar] [CrossRef] [PubMed]
  3. Salloum, G.; Bresnick, A.R.; Backer, J.M. Macropinocytosis: Mechanisms and regulation. Biochem. J. 2023, 480, 335–362. [Google Scholar] [CrossRef] [PubMed]
  4. Lambies, G.; Commisso, C. Macropinocytosis and Cancer: From Tumor Stress to Signaling Pathways. Subcell. Biochem. 2022, 98, 15–40. [Google Scholar] [CrossRef] [PubMed]
  5. Stow, J.L.; Hung, Y.; Wall, A.A. Macropinocytosis: Insights from immunology and cancer. Curr. Opin. Cell Biol. 2020, 65, 131–140. [Google Scholar] [CrossRef] [PubMed]
  6. Schink, K.O.; Tan, K.W.; Spangenberg, H.; Martorana, D.; Sneeggen, M.; Stevenin, V.; Enninga, J.; Campsteijn, C.; Raiborg, C.; Stenmark, H. The phosphoinositide coincidence detector Phafin2 promotes macropinocytosis by coordinating actin organisation at forming macropinosomes. Nat. Commun. 2021, 12, 6577. [Google Scholar] [CrossRef] [PubMed]
  7. 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]
  8. Mercer, J.; Helenius, A. Virus entry by macropinocytosis. Nat. Cell Biol. 2009, 11, 510–520. [Google Scholar] [CrossRef] [PubMed]
  9. Liu, H.; Qian, F. Exploiting macropinocytosis for drug delivery into KRAS mutant cancer. Theranostics 2022, 12, 1321–1332. [Google Scholar] [CrossRef] [PubMed]
  10. Egami, Y.; Taguchi, T.; Maekawa, M.; Arai, H.; Araki, N. Small GTPases and phosphoinositides in the regulatory mechanisms of macropinosome formation and maturation. Front. Physiol. 2014, 5, 374. [Google Scholar] [CrossRef]
  11. Walsh, A.B.; Bar-Sagi, D. Differential activation of the Rac pathway by Ha-Ras and K-Ras. J. Biol. Chem. 2001, 276, 15609–15615. [Google Scholar] [CrossRef] [PubMed]
  12. Hall, A. Ras-related GTPases and the cytoskeleton. Mol. Biol. Cell 1992, 3, 475–479. [Google Scholar] [CrossRef] [PubMed]
  13. 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]
  14. Kim, S.M.; Nguyen, T.T.; Ravi, A.; Kubiniok, P.; Finicle, B.T.; Jayashankar, V.; Malacrida, L.; Hou, J.; Robertson, J.; Gao, D.; et al. PTEN Deficiency and AMPK Activation Promote Nutrient Scavenging and Anabolism in Prostate Cancer Cells. Cancer Discov. 2018, 8, 866–883. [Google Scholar] [CrossRef]
  15. King, B.; Araki, J.; Palm, W.; Thompson, C.B. Yap/Taz promote the scavenging of extracellular nutrients through macropinocytosis. Genes Dev. 2020, 34, 1345–1358. [Google Scholar] [CrossRef]
  16. Lee, S.W.; Zhang, Y.; Jung, M.; Cruz, N.; Alas, B.; Commisso, C. EGFR-Pak Signaling Selectively Regulates Glutamine Deprivation-Induced Macropinocytosis. Dev. Cell 2019, 50, 381–392.e5. [Google Scholar] [CrossRef]
  17. Dowrick, P.; Kenworthy, P.; McCann, B.; Warn, R. Circular ruffle formation and closure lead to macropinocytosis in hepatocyte growth factor/scatter factor-treated cells. Eur. J. Cell Biol. 1993, 61, 44–53. [Google Scholar] [PubMed]
  18. Anton, I.M.; Saville, S.P.; Byrne, M.J.; Curcio, C.; Ramesh, N.; Hartwig, J.H.; Geha, R.S. WIP participates in actin reorganization and ruffle formation induced by PDGF. J. Cell Sci. 2003, 116, 2443–2451. [Google Scholar] [CrossRef]
  19. Racoosin, E.L.; Swanson, J.A. M-CSF-induced macropinocytosis increases solute endocytosis but not receptor-mediated endocytosis in mouse macrophages. J. Cell Sci. 1992, 102 Pt 4, 867–880. [Google Scholar] [CrossRef]
  20. Zdzalik-Bielecka, D.; Poswiata, A.; Kozik, K.; Jastrzebski, K.; Schink, K.O.; Brewinska-Olchowik, M.; Piwocka, K.; Stenmark, H.; Miaczynska, M. The GAS6-AXL signaling pathway triggers actin remodeling that drives membrane ruffling, macropinocytosis, and cancer-cell invasion. Proc. Natl. Acad. Sci. USA 2021, 118, e2024596118. [Google Scholar] [CrossRef]
  21. Tejeda-Munoz, N.; Albrecht, L.V.; Bui, M.H.; De Robertis, E.M. Wnt canonical pathway activates macropinocytosis and lysosomal degradation of extracellular proteins. Proc. Natl. Acad. Sci. USA 2019, 116, 10402–10411. [Google Scholar] [CrossRef] [PubMed]
  22. Redelman-Sidi, G.; Binyamin, A.; Gaeta, I.; Palm, W.; Thompson, C.B.; Romesser, P.B.; Lowe, S.W.; Bagul, M.; Doench, J.G.; Root, D.E.; et al. The Canonical Wnt Pathway Drives Macropinocytosis in Cancer. Cancer Res. 2018, 78, 4658–4670. [Google Scholar] [CrossRef] [PubMed]
  23. Sallusto, F.; Cella, M.; Danieli, C.; Lanzavecchia, A. Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromolecules in the major histocompatibility complex class II compartment: Downregulation by cytokines and bacterial products. J. Exp. Med. 1995, 182, 389–400. [Google Scholar] [CrossRef] [PubMed]
  24. Salloum, G.; Jakubik, C.T.; Erami, Z.; Heitz, S.D.; Bresnick, A.R.; Backer, J.M. PI3Kbeta is selectively required for growth factor-stimulated macropinocytosis. J. Cell Sci. 2019, 132, jcs231639. [Google Scholar] [CrossRef] [PubMed]
  25. Araki, N.; Johnson, M.T.; Swanson, J.A. A role for phosphoinositide 3-kinase in the completion of macropinocytosis and phagocytosis by macrophages. J. Cell Biol. 1996, 135, 1249–1260. [Google Scholar] [CrossRef] [PubMed]
  26. Ganot, P.; Tambutte, E.; Caminiti-Segonds, N.; Toullec, G.; Allemand, D.; Tambutte, S. Ubiquitous macropinocytosis in anthozoans. Elife 2020, 9, e50022. [Google Scholar] [CrossRef]
  27. Canton, J.; Schlam, D.; Breuer, C.; Gutschow, M.; Glogauer, M.; Grinstein, S. Calcium-sensing receptors signal constitutive macropinocytosis and facilitate the uptake of NOD2 ligands in macrophages. Nat. Commun. 2016, 7, 11284. [Google Scholar] [CrossRef]
  28. Mylvaganam, S.; Freeman, S.A.; Grinstein, S. The cytoskeleton in phagocytosis and macropinocytosis. Curr. Biol. 2021, 31, R619–R632. [Google Scholar] [CrossRef]
  29. Ha, K.D.; Bidlingmaier, S.M.; Liu, B. Macropinocytosis Exploitation by Cancers and Cancer Therapeutics. Front. Physiol. 2016, 7, 381. [Google Scholar] [CrossRef]
  30. Hoon, J.L.; Wong, W.K.; Koh, C.G. Functions and regulation of circular dorsal ruffles. Mol. Cell Biol. 2012, 32, 4246–4257. [Google Scholar] [CrossRef]
  31. Recouvreux, M.V.; Commisso, C. Macropinocytosis: A Metabolic Adaptation to Nutrient Stress in Cancer. Front. Endocrinol. 2017, 8, 261. [Google Scholar] [CrossRef] [PubMed]
  32. Bohdanowicz, M.; Schlam, D.; Hermansson, M.; Rizzuti, D.; Fairn, G.D.; Ueyama, T.; Somerharju, P.; Du, G.; Grinstein, S. Phosphatidic acid is required for the constitutive ruffling and macropinocytosis of phagocytes. Mol. Biol. Cell 2013, 24, 1700–1712, S1701–S1707. [Google Scholar] [CrossRef] [PubMed]
  33. Lanzetti, L.; Palamidessi, A.; Areces, L.; Scita, G.; Di Fiore, P.P. Rab5 is a signalling GTPase involved in actin remodelling by receptor tyrosine kinases. Nature 2004, 429, 309–314. [Google Scholar] [CrossRef] [PubMed]
  34. de Carvalho, T.M.; Barrias, E.S.; de Souza, W. Macropinocytosis: A pathway to protozoan infection. Front. Physiol. 2015, 6, 106. [Google Scholar] [CrossRef] [PubMed]
  35. Dharmawardhane, S.; Schurmann, A.; Sells, M.A.; Chernoff, J.; Schmid, S.L.; Bokoch, G.M. Regulation of macropinocytosis by p21-activated kinase-1. Mol. Biol. Cell 2000, 11, 3341–3352. [Google Scholar] [CrossRef] [PubMed]
  36. Fujii, M.; Kawai, K.; Egami, Y.; Araki, N. Dissecting the roles of Rac1 activation and deactivation in macropinocytosis using microscopic photo-manipulation. Sci. Rep. 2013, 3, 2385. [Google Scholar] [CrossRef] [PubMed]
  37. Ellerbroek, S.M.; Wennerberg, K.; Arthur, W.T.; Dunty, J.M.; Bowman, D.R.; DeMali, K.A.; Der, C.; Burridge, K. SGEF, a RhoG guanine nucleotide exchange factor that stimulates macropinocytosis. Mol. Biol. Cell 2004, 15, 3309–3319. [Google Scholar] [CrossRef] [PubMed]
  38. Katoh, H.; Negishi, M. RhoG activates Rac1 by direct interaction with the Dock180-binding protein Elmo. Nature 2003, 424, 461–464. [Google Scholar] [CrossRef] [PubMed]
  39. Samson, T.; Welch, C.; Monaghan-Benson, E.; Hahn, K.M.; Burridge, K. Endogenous RhoG is rapidly activated after epidermal growth factor stimulation through multiple guanine-nucleotide exchange factors. Mol. Biol. Cell 2010, 21, 1629–1642. [Google Scholar] [CrossRef] [PubMed]
  40. Zawistowski, J.S.; Sabouri-Ghomi, M.; Danuser, G.; Hahn, K.M.; Hodgson, L. A RhoC biosensor reveals differences in the activation kinetics of RhoA and RhoC in migrating cells. PLoS ONE 2013, 8, e79877. [Google Scholar] [CrossRef]
  41. Pertz, O.; Hodgson, L.; Klemke, R.L.; Hahn, K.M. Spatiotemporal dynamics of RhoA activity in migrating cells. Nature 2006, 440, 1069–1072. [Google Scholar] [CrossRef] [PubMed]
  42. Song, S.; Zhang, Y.; Ding, T.; Ji, N.; Zhao, H. The Dual Role of Macropinocytosis in Cancers: Promoting Growth and Inducing Methuosis to Participate in Anticancer Therapies as Targets. Front. Oncol. 2020, 10, 570108. [Google Scholar] [CrossRef] [PubMed]
  43. Sun, P.; Yamamoto, H.; Suetsugu, S.; Miki, H.; Takenawa, T.; Endo, T. Small GTPase Rah/Rab34 is associated with membrane ruffles and macropinosomes and promotes macropinosome formation. J. Biol. Chem. 2003, 278, 4063–4071. [Google Scholar] [CrossRef] [PubMed]
  44. Sun, P.; Endo, T. Assays for functional properties of Rab34 in macropinosome formation. Methods Enzymol. 2005, 403, 229–243. [Google Scholar] [CrossRef] [PubMed]
  45. Coyne, C.B.; Shen, L.; Turner, J.R.; Bergelson, J.M. Coxsackievirus entry across epithelial tight junctions requires occludin and the small GTPases Rab34 and Rab5. Cell Host Microbe 2007, 2, 181–192. [Google Scholar] [CrossRef] [PubMed]
  46. Welliver, T.P.; Swanson, J.A. A growth factor signaling cascade confined to circular ruffles in macrophages. Biol. Open 2012, 1, 754–760. [Google Scholar] [CrossRef] [PubMed]
  47. Honda, A.; Nogami, M.; Yokozeki, T.; Yamazaki, M.; Nakamura, H.; Watanabe, H.; Kawamoto, K.; Nakayama, K.; Morris, A.J.; Frohman, M.A.; et al. Phosphatidylinositol 4-phosphate 5-kinase alpha is a downstream effector of the small G protein ARF6 in membrane ruffle formation. Cell 1999, 99, 521–532. [Google Scholar] [CrossRef] [PubMed]
  48. Feliciano, W.D.; Yoshida, S.; Straight, S.W.; Swanson, J.A. Coordination of the Rab5 cycle on macropinosomes. Traffic 2011, 12, 1911–1922. [Google Scholar] [CrossRef] [PubMed]
  49. Lim, J.P.; Gleeson, P.A. Macropinocytosis: An endocytic pathway for internalising large gulps. Immunol. Cell Biol. 2011, 89, 836–843. [Google Scholar] [CrossRef]
  50. Zeziulia, M.; Blin, S.; Schmitt, F.W.; Lehmann, M.; Jentsch, T.J. Proton-gated anion transport governs macropinosome shrinkage. Nat. Cell Biol. 2022, 24, 885–895. [Google Scholar] [CrossRef]
  51. Wang, X.; Zhang, X.; Dong, X.P.; Samie, M.; Li, X.; Cheng, X.; Goschka, A.; Shen, D.; Zhou, Y.; Harlow, J.; et al. TPC proteins are phosphoinositide- activated sodium-selective ion channels in endosomes and lysosomes. Cell 2012, 151, 372–383. [Google Scholar] [CrossRef]
  52. Krishna, S.; Palm, W.; Lee, Y.; Yang, W.; Bandyopadhyay, U.; Xu, H.; Florey, O.; Thompson, C.B.; Overholtzer, M. PIKfyve Regulates Vacuole Maturation and Nutrient Recovery following Engulfment. Dev. Cell 2016, 38, 536–547. [Google Scholar] [CrossRef]
  53. Cang, C.; Zhou, Y.; Navarro, B.; Seo, Y.J.; Aranda, K.; Shi, L.; Battaglia-Hsu, S.; Nissim, I.; Clapham, D.E.; Ren, D. mTOR regulates lysosomal ATP-sensitive two-pore Na(+) channels to adapt to metabolic state. Cell 2013, 152, 778–790. [Google Scholar] [CrossRef]
  54. Wang, J.T.; Kerr, M.C.; Karunaratne, S.; Jeanes, A.; Yap, A.S.; Teasdale, R.D. The SNX-PX-BAR family in macropinocytosis: The regulation of macropinosome formation by SNX-PX-BAR proteins. PLoS ONE 2010, 5, e13763. [Google Scholar] [CrossRef]
  55. Zerial, M.; McBride, H. Rab proteins as membrane organizers. Nat. Rev. Mol. Cell Biol. 2001, 2, 107–117. [Google Scholar] [CrossRef]
  56. Wandinger-Ness, A.; Zerial, M. Rab proteins and the compartmentalization of the endosomal system. Cold Spring Harb. Perspect. Biol. 2014, 6, a022616. [Google Scholar] [CrossRef]
  57. Morishita, S.; Wada, N.; Fukuda, M.; Nakamura, T. Rab5 activation on macropinosomes requires ALS2, and subsequent Rab5 inactivation through ALS2 detachment requires active Rab7. FEBS Lett. 2019, 593, 230–241. [Google Scholar] [CrossRef]
  58. Liu, Z.; Xu, E.; Zhao, H.T.; Cole, T.; West, A.B. LRRK2 and Rab10 coordinate macropinocytosis to mediate immunological responses in phagocytes. EMBO J. 2020, 39, e104862. [Google Scholar] [CrossRef]
  59. Egami, Y.; Araki, N. Spatiotemporal Localization of Rab20 in Live RAW264 Macrophages during Macropinocytosis. Acta Histochem. Cytochem. 2012, 45, 317–323. [Google Scholar] [CrossRef]
  60. Egami, Y.; Araki, N. Dynamic changes in the spatiotemporal localization of Rab21 in live RAW264 cells during macropinocytosis. PLoS ONE 2009, 4, e6689. [Google Scholar] [CrossRef]
  61. Jiang, G.; Wei, C.; Chen, Y.; Lyu, Y.; Huang, J.; Chen, H.; Gao, X. Targeted drug delivery system inspired by macropinocytosis. J. Control Release 2023, 359, 302–314. [Google Scholar] [CrossRef]
  62. Donaldson, J.G. Macropinosome formation, maturation and membrane recycling: Lessons from clathrin-independent endosomal membrane systems. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2019, 374, 20180148. [Google Scholar] [CrossRef]
  63. O'Sullivan, M.J.; Lindsay, A.J. The Endosomal Recycling Pathway-At the Crossroads of the Cell. Int. J. Mol. Sci. 2020, 21, 6074. [Google Scholar] [CrossRef]
  64. Wolpin, B.M.; Rubinson, D.A.; Wang, X.; Chan, J.A.; Cleary, J.M.; Enzinger, P.C.; Fuchs, C.S.; McCleary, N.J.; Meyerhardt, J.A.; Ng, K.; et al. Phase II and pharmacodynamic study of autophagy inhibition using hydroxychloroquine in patients with metastatic pancreatic adenocarcinoma. Oncologist 2014, 19, 637–638. [Google Scholar] [CrossRef]
  65. Hewlett, L.J.; Prescott, A.R.; Watts, C. The coated pit and macropinocytic pathways serve distinct endosome populations. J. Cell Biol. 1994, 124, 689–703. [Google Scholar] [CrossRef]
  66. Yoshimori, T.; Yamamoto, A.; Moriyama, Y.; Futai, M.; Tashiro, Y. Bafilomycin A1, a specific inhibitor of vacuolar-type H(+)-ATPase, inhibits acidification and protein degradation in lysosomes of cultured cells. J. Biol. Chem. 1991, 266, 17707–17712. [Google Scholar] [CrossRef]
  67. Shubin, A.V.; Demidyuk, I.V.; Lunina, N.A.; Komissarov, A.A.; Roschina, M.P.; Leonova, O.G.; Kostrov, S.V. Protease 3C of hepatitis A virus induces vacuolization of lysosomal/endosomal organelles and caspase-independent cell death. BMC Cell Biol. 2015, 16, 4. [Google Scholar] [CrossRef]
  68. Cotter, K.; Capecci, J.; Sennoune, S.; Huss, M.; Maier, M.; Martinez-Zaguilan, R.; Forgac, M. Activity of plasma membrane V-ATPases is critical for the invasion of MDA-MB231 breast cancer cells. J. Biol. Chem. 2015, 290, 3680–3692. [Google Scholar] [CrossRef]
  69. Chung, C.; Mader, C.C.; Schmitz, J.C.; Atladottir, J.; Fitchev, P.; Cornwell, M.L.; Koleske, A.J.; Crawford, S.E.; Gorelick, F. The vacuolar-ATPase modulates matrix metalloproteinase isoforms in human pancreatic cancer. Lab. Investig. 2011, 91, 732–743. [Google Scholar] [CrossRef]
  70. Ritter, M.; Bresgen, N.; Kerschbaum, H.H. From Pinocytosis to Methuosis-Fluid Consumption as a Risk Factor for Cell Death. Front. Cell Dev. Biol. 2021, 9, 651982. [Google Scholar] [CrossRef]
  71. Buckley, C.M.; King, J.S. Drinking problems: Mechanisms of macropinosome formation and maturation. FEBS J. 2017, 284, 3778–3790. [Google Scholar] [CrossRef]
  72. Qiu, Z.; Liu, W.; Zhu, Q.; Ke, K.; Zhu, Q.; Jin, W.; Yu, S.; Yang, Z.; Li, L.; Sun, X.; et al. The Role and Therapeutic Potential of Macropinocytosis in Cancer. Front. Pharmacol. 2022, 13, 919819. [Google Scholar] [CrossRef]
  73. Shubin, A.V.; Demidyuk, I.V.; Komissarov, A.A.; Rafieva, L.M.; Kostrov, S.V. Cytoplasmic vacuolization in cell death and survival. Oncotarget 2016, 7, 55863–55889. [Google Scholar] [CrossRef]
  74. Overmeyer, J.H.; Kaul, A.; Johnson, E.E.; Maltese, W.A. Active ras triggers death in glioblastoma cells through hyperstimulation of macropinocytosis. Mol. Cancer Res. 2008, 6, 965–977. [Google Scholar] [CrossRef]
  75. Maltese, W.A.; Overmeyer, J.H. Methuosis: Nonapoptotic cell death associated with vacuolization of macropinosome and endosome compartments. Am. J. Pathol. 2014, 184, 1630–1642. [Google Scholar] [CrossRef]
  76. Gao, X.; Ji, C.; Wang, J.; Song, X.; Zuo, R.; Zhang, J.; Chen, X.; Ji, H.; Peng, L.; Guo, D.; et al. Maduramicin induces cardiotoxicity via Rac1 signaling-independent methuosis in H9c2 cells. J. Appl. Toxicol. 2021, 41, 1937–1951. [Google Scholar] [CrossRef]
  77. Kaul, A.; Overmeyer, J.H.; Maltese, W.A. Activated Ras induces cytoplasmic vacuolation and non-apoptotic death in glioblastoma cells via novel effector pathways. Cell Signal 2007, 19, 1034–1043. [Google Scholar] [CrossRef]
  78. Bhanot, H.; Young, A.M.; Overmeyer, J.H.; Maltese, W.A. Induction of nonapoptotic cell death by activated Ras requires inverse regulation of Rac1 and Arf6. Mol. Cancer Res. 2010, 8, 1358–1374. [Google Scholar] [CrossRef]
  79. Manara, M.C.; Terracciano, M.; Mancarella, C.; Sciandra, M.; Guerzoni, C.; Pasello, M.; Grilli, A.; Zini, N.; Picci, P.; Colombo, M.P.; et al. CD99 triggering induces methuosis of Ewing sarcoma cells through IGF-1R/RAS/Rac1 signaling. Oncotarget 2016, 7, 79925–79942. [Google Scholar] [CrossRef]
  80. Li, Z.; Mbah, N.E.; Overmeyer, J.H.; Sarver, J.G.; George, S.; Trabbic, C.J.; Erhardt, P.W.; Maltese, W.A. The JNK signaling pathway plays a key role in methuosis (non-apoptotic cell death) induced by MOMIPP in glioblastoma. BMC Cancer 2019, 19, 77. [Google Scholar] [CrossRef]
  81. Cho, H.; Geno, E.; Patoor, M.; Reid, A.; McDonald, R.; Hild, M.; Jenkins, J.L. Indolyl-Pyridinyl-Propenone-Induced Methuosis through the Inhibition of PIKFYVE. ACS Omega 2018, 3, 6097–6103. [Google Scholar] [CrossRef]
  82. de Lartigue, J.; Polson, H.; Feldman, M.; Shokat, K.; Tooze, S.A.; Urbe, S.; Clague, M.J. PIKfyve regulation of endosome-linked pathways. Traffic 2009, 10, 883–893. [Google Scholar] [CrossRef]
  83. Shisheva, A. PIKfyve: Partners, significance, debates and paradoxes. Cell Biol. Int. 2008, 32, 591–604. [Google Scholar] [CrossRef]
  84. Li, R.; Ng, T.S.C.; Wang, S.J.; Prytyskach, M.; Rodell, C.B.; Mikula, H.; Kohler, R.H.; Garlin, M.A.; Lauffenburger, D.A.; Parangi, S.; et al. Therapeutically reprogrammed nutrient signalling enhances nanoparticulate albumin bound drug uptake and efficacy in KRAS-mutant cancer. Nat. Nanotechnol. 2021, 16, 830–839. [Google Scholar] [CrossRef]
  85. Liu, H.; Sun, M.; Liu, Z.; Kong, C.; Kong, W.; Ye, J.; Gong, J.; Huang, D.C.S.; Qian, F. KRAS-enhanced macropinocytosis and reduced FcRn-mediated recycling sensitize pancreatic cancer to albumin-conjugated drugs. J. Control Release 2019, 296, 40–53. [Google Scholar] [CrossRef]
  86. Gong, X.; Sun, R.; Gao, Z.; Han, W.; Liu, Y.; Zhao, L.; Jing, L.; Yao, X.; Sun, X. Tubeimoside 1 Acts as a Chemotherapeutic Synergist via Stimulating Macropinocytosis. Front. Pharmacol. 2018, 9, 1044. [Google Scholar] [CrossRef]
  87. Silva-Pavez, E.; Villar, P.; Trigo, C.; Caamano, E.; Niechi, I.; Perez, P.; Munoz, J.P.; Aguayo, F.; Burzio, V.A.; Varas-Godoy, M.; et al. CK2 inhibition with silmitasertib promotes methuosis-like cell death associated to catastrophic massive vacuolization of colorectal cancer cells. Cell Death Dis. 2019, 10, 73. [Google Scholar] [CrossRef]
  88. Sarkar Bhattacharya, S.; Thirusangu, P.; Jin, L.; Roy, D.; Jung, D.; Xiao, Y.; Staub, J.; Roy, B.; Molina, J.R.; Shridhar, V. PFKFB3 inhibition reprograms malignant pleural mesothelioma to nutrient stress-induced macropinocytosis and ER stress as independent binary adaptive responses. Cell Death Dis. 2019, 10, 725. [Google Scholar] [CrossRef]
  89. Su, H.; Yang, F.; Fu, R.; Li, X.; French, R.; Mose, E.; Pu, X.; Trinh, B.; Kumar, A.; Liu, J.; et al. Cancer cells escape autophagy inhibition via NRF2-induced macropinocytosis. Cancer Cell 2021, 39, 678–693.e611. [Google Scholar] [CrossRef]
  90. Ramirez, C.; Hauser, A.D.; Vucic, E.A.; Bar-Sagi, D. Plasma membrane V-ATPase controls oncogenic RAS-induced macropinocytosis. Nature 2019, 576, 477–481. [Google Scholar] [CrossRef]
  91. Thu, P.M.; Zheng, Z.G.; Zhou, Y.P.; Wang, Y.Y.; Zhang, X.; Jing, D.; Cheng, H.M.; Li, J.; Li, P.; Xu, X. Phellodendrine chloride suppresses proliferation of KRAS mutated pancreatic cancer cells through inhibition of nutrients uptake via macropinocytosis. Eur. J. Pharmacol. 2019, 850, 23–34. [Google Scholar] [CrossRef]
  92. Xiao, F.; Li, J.; Huang, K.; Li, X.; Xiong, Y.; Wu, M.; Wu, L.; Kuang, W.; Lv, S.; Wu, L.; et al. Macropinocytosis: Mechanism and targeted therapy in cancers. Am. J. Cancer Res. 2021, 11, 14–30. [Google Scholar]
  93. Adkins, D.; Ley, J.; Atiq, O.; Powell, S.; Spanos, W.C.; Gitau, M.; Rigden, C.; Palka, K.; Liu, J.; Oppelt, P. Nanoparticle albumin-bound paclitaxel with cetuximab and carboplatin as first-line therapy for recurrent or metastatic head and neck cancer: A single-arm, multicenter, phase 2 trial. Oral. Oncol. 2021, 115, 105173. [Google Scholar] [CrossRef]
  94. Du, Y.; Shang, B.Y.; Sheng, W.J.; Zhang, S.H.; Li, Y.; Miao, Q.F.; Zhen, Y.S. A recombinantly tailored beta-defensin that displays intensive macropinocytosis-mediated uptake exerting potent efficacy against K-Ras mutant pancreatic cancer. Oncotarget 2016, 7, 58418–58434. [Google Scholar] [CrossRef]
  95. Takenaka, T.; Nakai, S.; Katayama, M.; Hirano, M.; Ueno, N.; Noguchi, K.; Takatani-Nakase, T.; Fujii, I.; Kobayashi, S.S.; Nakase, I. Effects of gefitinib treatment on cellular uptake of extracellular vesicles in EGFR-mutant non-small cell lung cancer cells. Int. J. Pharm. 2019, 572, 118762. [Google Scholar] [CrossRef]
  96. Kong, C.; Li, Y.; Liu, Z.; Ye, J.; Wang, Z.; Zhang, L.; Kong, W.; Liu, H.; Liu, C.; Pang, H.; et al. Targeting the Oncogene KRAS Mutant Pancreatic Cancer by Synergistic Blocking of Lysosomal Acidification and Rapid Drug Release. ACS Nano 2019, 13, 4049–4063. [Google Scholar] [CrossRef]
  97. Kitazawa, S.; Nishizawa, S.; Nakagawa, H.; Funata, M.; Nishimura, K.; Soga, T.; Hara, T. Cancer with low cathepsin D levels is susceptible to vacuolar (H(+) )-ATPase inhibition. Cancer Sci. 2017, 108, 1185–1193. [Google Scholar] [CrossRef]
  98. Wang, T.; Zhang, Y.; Liu, Y.; Huang, Y.; Wang, W. Amino Acid-Starved Cancer Cells Utilize Macropinocytosis and Ubiquitin-Proteasome System for Nutrient Acquisition. Adv. Sci. 2024, 11, e2304791. [Google Scholar] [CrossRef]
  99. Tajiri, H.; Uruno, T.; Shirai, T.; Takaya, D.; Matsunaga, S.; Setoyama, D.; Watanabe, M.; Kukimoto-Niino, M.; Oisaki, K.; Ushijima, M.; et al. Targeting Ras-Driven Cancer Cell Survival and Invasion through Selective Inhibition of DOCK1. Cell Rep. 2017, 19, 969–980. [Google Scholar] [CrossRef]
  100. Srivastava, R.K.; Li, C.; Khan, J.; Banerjee, N.S.; Chow, L.T.; Athar, M. Combined mTORC1/mTORC2 inhibition blocks growth and induces catastrophic macropinocytosis in cancer cells. Proc. Natl. Acad. Sci. USA 2019, 116, 24583–24592. [Google Scholar] [CrossRef]
  101. Palm, W.; Park, Y.; Wright, K.; Pavlova, N.N.; Tuveson, D.A.; Thompson, C.B. The Utilization of Extracellular Proteins as Nutrients Is Suppressed by mTORC1. Cell 2015, 162, 259–270. [Google Scholar] [CrossRef]
  102. Michalopoulou, E.; Auciello, F.R.; Bulusu, V.; Strachan, D.; Campbell, A.D.; Tait-Mulder, J.; Karim, S.A.; Morton, J.P.; Sansom, O.J.; Kamphorst, J.J. Macropinocytosis Renders a Subset of Pancreatic Tumor Cells Resistant to mTOR Inhibition. Cell Rep. 2020, 30, 2729–2742.e2724. [Google Scholar] [CrossRef]
  103. Song, S.; Xia, X.; Qi, J.; Hu, X.; Chen, Q.; Liu, J.; Ji, N.; Zhao, H. Silmitasertib-induced macropinocytosis promoting DDP intracellular uptake to enhance cell apoptosis in oral squamous cell carcinoma. Drug Deliv. 2021, 28, 2480–2494. [Google Scholar] [CrossRef]
  104. Sun, L.; Li, B.; Su, X.; Chen, G.; Li, Y.; Yu, L.; Li, L.; Wei, W. An Ursolic Acid Derived Small Molecule Triggers Cancer Cell Death through Hyperstimulation of Macropinocytosis. J. Med. Chem. 2017, 60, 6638–6648. [Google Scholar] [CrossRef]
  105. Ahlstedt, J.; Fornvik, K.; Zolfaghari, S.; Kwak, D.; Hammarstrom, L.G.J.; Ernfors, P.; Salford, L.G.; Redebrandt, H.N. Evaluating vacquinol-1 in rats carrying glioblastoma models RG2 and NS1. Oncotarget 2018, 9, 8391–8399. [Google Scholar] [CrossRef]
  106. Jeon, D.; Kim, S.W.; Kim, H.S. Platycodin D, a bioactive component of Platycodon grandiflorum, induces cancer cell death associated with extreme vacuolation. Anim. Cells Syst. 2019, 23, 118–127. [Google Scholar] [CrossRef]
  107. Cingolani, F.; Simbari, F.; Abad, J.L.; Casasampere, M.; Fabrias, G.; Futerman, A.H.; Casas, J. Jaspine B induces nonapoptotic cell death in gastric cancer cells independently of its inhibition of ceramide synthase. J. Lipid Res. 2017, 58, 1500–1513. [Google Scholar] [CrossRef]
  108. Liu, X.; Wang, S.; Zheng, H.; Liu, Q.; Shen, T.; Wang, X.; Ren, D. Epimedokoreanin C, a prenylated flavonoid isolated from Epimedium koreanum, induces non-apoptotic cell death with the characteristics of methuosis in lung cancer cells. Am. J. Cancer Res. 2021, 11, 3496–3514. [Google Scholar]
  109. Kimiz-Gebologlu, I.; Gulce-Iz, S.; Biray-Avci, C. Monoclonal antibodies in cancer immunotherapy. Mol. Biol. Rep. 2018, 45, 2935–2940. [Google Scholar] [CrossRef]
  110. Wang, X.; Sheng, W.; Wang, Y.; Li, L.; Li, Y.; Zhang, S.; Liu, X.; Chen, S.; Zhen, Y. A Macropinocytosis-Intensifying Albumin Domain-Based scFv Antibody and Its Conjugate Directed against K-Ras Mutant Pancreatic Cancer. Mol. Pharm. 2018, 15, 2403–2412. [Google Scholar] [CrossRef]
  111. Zhao, H.; Atkinson, J.; Gulesserian, S.; Zeng, Z.; Nater, J.; Ou, J.; Yang, P.; Morrison, K.; Coleman, J.; Malik, F.; et al. Modulation of Macropinocytosis-Mediated Internalization Decreases Ocular Toxicity of Antibody-Drug Conjugates. Cancer Res. 2018, 78, 2115–2126. [Google Scholar] [CrossRef]
  112. Redelman-Sidi, G.; Iyer, G.; Solit, D.B.; Glickman, M.S. Oncogenic activation of Pak1-dependent pathway of macropinocytosis determines BCG entry into bladder cancer cells. Cancer Res. 2013, 73, 1156–1167. [Google Scholar] [CrossRef] [PubMed]
  113. Alvarez-Arguedas, S.; Uranga, S.; Martin, M.; Elizalde, J.; Gomez, A.B.; Julian, E.; Nardelli-Haefliger, D.; Martin, C.; Aguilo, N. Therapeutic efficacy of the live-attenuated Mycobacterium tuberculosis vaccine, MTBVAC, in a preclinical model of bladder cancer. Transl. Res. 2018, 197, 32–42. [Google Scholar] [CrossRef]
  114. Zhou, S.; Huang, Y.; Chen, Y.; Liu, S.; Xu, M.; Jiang, T.; Song, Q.; Jiang, G.; Gu, X.; Gao, X.; et al. Engineering ApoE3-incorporated biomimetic nanoparticle for efficient vaccine delivery to dendritic cells via macropinocytosis to enhance cancer immunotherapy. Biomaterials 2020, 235, 119795. [Google Scholar] [CrossRef]
  115. Huang, J.L.; Jiang, G.; Song, Q.X.; Gu, X.; Hu, M.; Wang, X.L.; Song, H.H.; Chen, L.P.; Lin, Y.Y.; Jiang, D.; et al. Lipoprotein-biomimetic nanostructure enables efficient targeting delivery of siRNA to Ras-activated glioblastoma cells via macropinocytosis. Nat. Commun. 2017, 8, 15144. [Google Scholar] [CrossRef]
  116. Lu, M.; Zhao, X.; Xing, H.; Xun, Z.; Zhu, S.; Lang, L.; Yang, T.; Cai, C.; Wang, D.; Ding, P. Comparison of exosome-mimicking liposomes with conventional liposomes for intracellular delivery of siRNA. Int. J. Pharm. 2018, 550, 100–113. [Google Scholar] [CrossRef]
  117. Kamerkar, S.; LeBleu, V.S.; Sugimoto, H.; Yang, S.; Ruivo, C.F.; Melo, S.A.; Lee, J.J.; Kalluri, R. Exosomes facilitate therapeutic targeting of oncogenic KRAS in pancreatic cancer. Nature 2017, 546, 498–503. [Google Scholar] [CrossRef]
  118. Jiang, G.; Chen, H.; Huang, J.; Song, Q.; Chen, Y.; Gu, X.; Jiang, Z.; Huang, Y.; Lin, Y.; Feng, J.; et al. Tailored Lipoprotein-Like miRNA Delivery Nanostructure Suppresses Glioma Stemness and Drug Resistance through Receptor-Stimulated Macropinocytosis. Adv. Sci. 2020, 7, 1903290. [Google Scholar] [CrossRef]
  119. Karaki, S.; Benizri, S.; Mejias, R.; Baylot, V.; Branger, N.; Nguyen, T.; Vialet, B.; Oumzil, K.; Barthelemy, P.; Rocchi, P. Lipid-oligonucleotide conjugates improve cellular uptake and efficiency of TCTP-antisense in castration-resistant prostate cancer. J. Control Release 2017, 258, 1–9. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Cellular process and regulation of macropinocytosis. As shown in the colored areas, macropinocytic process is divided into 4 stages: (1) initiation and activation of macropinocytosis; (2) formation of macropinosomes; (3) maturation of macropinosomes; and (4) degradation and recycling of macropinosomes. Macropinocytosis is activated by a number of extracellular signals, such as growth factors and stress factors like amino acid deficiency and hypoxia. These signals either directly or indirectly activate RAS/RAC1/PI3K signaling to initiate plasma membrane ruffle formation. The formation of macropinosomes is involved in actin cytoskeleton remodeling followed by activation of RAS/RAC1/PI3K signaling. Multiple RAB proteins, such as RAB5, RAB20, RAB21, and RAB34, and ARF proteins, such as ARF6 and ARF1, also participate in the process along with the RAS/RAC1/PI3K proteins. The maturation of macropinosomes is osmatically processed by ion channels, such as TPC for Na+, 2Cl /H+ exchanger ClC5, and Cl channel TMEM206, for adjusting the volume of macropinosomes. The matured macropinosomes are transported to and fused with lysosomes for degradation. The fusion of matured macropinosomes with lysosomes is promoted by TRPML1, and degradation of macropinosomes is inhibited by mTORC1. Some macropinosomes can be recycled back to plasma membrane, and the recycling process is regulated by EHBP1L1 and ARF6. When macropinocytosis is over-activated and impaired, excessive macropinosomes can undergo vacuolization by self-fusion and form large dysfunctional vacuoles, leading to the non-apoptotic cell death called methuosis. The RAS/RAC1/PI3K proteins are the core regulatory proteins in macropinocytosis and their main function is generating plasma membrane ruffles, which is the essential cellular process for formation of macropinosomes. The major regulatory pathways in each of the process stages are labelled in the figure and described in the text. PI lipids stands for phsphatidylinositol lipids, including PI(3,4)P2, PI(4,5)P2, and PI(3,4,5)P3. PI(3,4)P2, phosphatidylinositol 3,4-bisphosphate; PI(4,5)P2, phosphatidylinositol 4,5-bisphosphate; PI(3,4,5)P3, phosphatidylinositol-3,4,5-trisphosphate; EV, extracellular vesicle; MPS, macropinosome; RTK, receptor tyrosine kinase.
Figure 1. Cellular process and regulation of macropinocytosis. As shown in the colored areas, macropinocytic process is divided into 4 stages: (1) initiation and activation of macropinocytosis; (2) formation of macropinosomes; (3) maturation of macropinosomes; and (4) degradation and recycling of macropinosomes. Macropinocytosis is activated by a number of extracellular signals, such as growth factors and stress factors like amino acid deficiency and hypoxia. These signals either directly or indirectly activate RAS/RAC1/PI3K signaling to initiate plasma membrane ruffle formation. The formation of macropinosomes is involved in actin cytoskeleton remodeling followed by activation of RAS/RAC1/PI3K signaling. Multiple RAB proteins, such as RAB5, RAB20, RAB21, and RAB34, and ARF proteins, such as ARF6 and ARF1, also participate in the process along with the RAS/RAC1/PI3K proteins. The maturation of macropinosomes is osmatically processed by ion channels, such as TPC for Na+, 2Cl /H+ exchanger ClC5, and Cl channel TMEM206, for adjusting the volume of macropinosomes. The matured macropinosomes are transported to and fused with lysosomes for degradation. The fusion of matured macropinosomes with lysosomes is promoted by TRPML1, and degradation of macropinosomes is inhibited by mTORC1. Some macropinosomes can be recycled back to plasma membrane, and the recycling process is regulated by EHBP1L1 and ARF6. When macropinocytosis is over-activated and impaired, excessive macropinosomes can undergo vacuolization by self-fusion and form large dysfunctional vacuoles, leading to the non-apoptotic cell death called methuosis. The RAS/RAC1/PI3K proteins are the core regulatory proteins in macropinocytosis and their main function is generating plasma membrane ruffles, which is the essential cellular process for formation of macropinosomes. The major regulatory pathways in each of the process stages are labelled in the figure and described in the text. PI lipids stands for phsphatidylinositol lipids, including PI(3,4)P2, PI(4,5)P2, and PI(3,4,5)P3. PI(3,4)P2, phosphatidylinositol 3,4-bisphosphate; PI(4,5)P2, phosphatidylinositol 4,5-bisphosphate; PI(3,4,5)P3, phosphatidylinositol-3,4,5-trisphosphate; EV, extracellular vesicle; MPS, macropinosome; RTK, receptor tyrosine kinase.
Ijms 25 06963 g001
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

Wu, Y.; Hu, X.; Wei, Z.; Lin, Q. Cellular Regulation of Macropinocytosis. Int. J. Mol. Sci. 2024, 25, 6963. https://doi.org/10.3390/ijms25136963

AMA Style

Wu Y, Hu X, Wei Z, Lin Q. Cellular Regulation of Macropinocytosis. International Journal of Molecular Sciences. 2024; 25(13):6963. https://doi.org/10.3390/ijms25136963

Chicago/Turabian Style

Wu, Yumeng, Xiao Hu, Zhixiao Wei, and Qiong Lin. 2024. "Cellular Regulation of Macropinocytosis" International Journal of Molecular Sciences 25, no. 13: 6963. https://doi.org/10.3390/ijms25136963

APA Style

Wu, Y., Hu, X., Wei, Z., & Lin, Q. (2024). Cellular Regulation of Macropinocytosis. International Journal of Molecular Sciences, 25(13), 6963. https://doi.org/10.3390/ijms25136963

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