Preprint
Review

This version is not peer-reviewed.

Molecular Detection of Membrane Damage: Lessons from Bacteria

Submitted:

26 September 2025

Posted:

28 September 2025

You are already at the latest version

Abstract

Mammalian cells contain many membranous organelles, among which endosomes are the initial destination for endocytosed materials. Drugs and pathogens, such as bacteria, are internalized by cells and transported to endosomes or phagosomes, then to lysosomes for degradation. Internalized drugs must escape from endosomes into the cytosol before undergoing degradation in lysosomes. However, endosomal escape is often inefficient in artificial drug delivery systems (DDSs). In contrast, many pathogens are phagocytosed and subsequently escape into the cytosol to proliferate. The studies on phagosomal escape of pathogens have revealed the molecular mechanisms through which host cells detect organelle membrane damage. In this review, we first provide an overview of bacterial endosomal and phagosomal escape, focusing on Shigella flexneri as a model organism. We then describe the current knowledge on the cellular machinery involved in sensing and repairing membrane damage, including galectins, ESCRTs, sphingomyelin, stress granules, PI4P in membrane contact sites, and Annexins. We further discuss the roles of secretory MVBs in plasma membrane repair in the Annexins and Future Perspectives sections. Research on membrane damage not only advances our understanding of cellular responses to damage caused by pathogens and artificial nanoparticles, but also informs the design of more effective DDSs.

Keywords: 
;  ;  ;  ;  ;  

1. Introduction

Mammalian cells internalize nutrients, solutes, growth factors, or hormones that bind to specific receptors on the plasma membrane through endocytosis. Early endosomes are the first organelles that internalized materials reach, which sort and transport these materials to their respective destinations. The endocytic pathways have been well reviewed elsewhere [1,2,3,4]. Briefly, after internalization, cargo is sorted either to lysosomes for degradation, or to early or recycling endosomes for transporting back to the plasma membrane. There is a retrograde transport route from early endosomes to the Golgi apparatus, which leads to the endoplasmic reticulum (ER).
Internalized materials first reach early endosomes with a pH of 6.0-6.3, then are further acidified to a pH of 5.3 in late endosomes; subsequently, proteins are degraded in lysosomes with a pH of 5.2-5.3 [5,6,7]. The bulk flow route is thought to be the degradation pathway, as maturation of early endosomes to late endosomes occurs rather than direct transport from pre-existing early endosomes to late endosomes [3,8]. However, transmembrane receptors and lipids undergo cargo sorting processes in early endosomes and can be transported to recycling pathways leading to the plasma membrane, the Golgi apparatus, or the degradation pathway [9,10]. In addition, there are other uncharacterized endosomal pathways [11,12,13].
In this review, we first overview Shigella flexneri, a well-studied bacterium that escapes from endosomes; then, we focus on molecular machineries in mammalian cells. After endosomal escape, autophagy is crucial for preventing Shigella proliferation [14]. Autophagy is a process in which cytosolic components are degraded after being sequestered within a double-membrane structure marked by lipidated LC3 [15,16]. However, in this review, we do not discuss autophagy in detail, as it has already been extensively covered in other articles. Moreover, multiple pathways can induce autophagy (see , 3-2, 3-3, and 3-5). Instead, we focus on the earlier phase of damaged organelle detection rather than on downstream autophagy process.

2. Phagosomal Escape by Shigella Species

2.1. Overview of Infection by Shigella spp.

Shigella species are Gram-negative bacteria belonging to the Enterobacteria that cause intestinal infection [17,18]. Shigella was first discovered by Dr. Kiyoshi Shiga in 1897 as the cause of epidemic dysentery, later named S. dysenteriae [19]. The genus Shigella comprises S. dysenteriae, S. flexneri, S. boydii, and S. sonnei. Among them, S. flexneri is the most extensively studied. M cells, specialized epithelial cells that continuously uptake gut antigens and transfer them to the associated lymphoid tissue, Peyer’s patches via transcytosis, have been identified as the initial uptake site of S. flexneri [20,21]. S. flexneri crosses the epithelial layer via transcytosis of M cells, then kills macrophages that have engulfed S. flexneri [22]. S. flexneri, which harbors a 220 kb plasmid, kills macrophages via apoptosis, but non-invasive S. flexneri strains or Listeria monocytogenes—a Gram-positive bacterium that invades the cellular cytoplasmdo not cause apoptosis. Therefore, the invasion of S. flexneri is important for macrophage apoptosis, and not all invasive bacteria induce apoptosis. After escaping from macrophages, S. flexneri is internalized by epithelial cells (ECs) and proliferates within them [23,24,25]. In experiments, S. flexneri is phagocytosed by non-professional phagocytes, such as Henle epithelial cells or HeLa cells, and this process is blocked by phagocytosis inhibitors [24]. S. flexneri utilizes the actin cytoskeleton of ECs to move [26] and invades adjacent ECs to evade immune responses outside the cells [27].
The invasion activity is encoded in a 30-37 kb region of the 220 kb plasmid of S. flexneri [28,29,30]. This region encodes the Type III secretion system (T3SS) and its effector proteins [17]. The T3SS comprises about 20 proteins forming a needle-like structure that crosses from the bacterial surface, including the peptidoglycan layer, to the host membrane [18,31]. Effector proteins are secreted by the T3SS to manipulate host cell activities.
Shiga toxin is secreted into the culture medium by S. dysenteriae [32], as well as by S. flexneri and S. sonnei in smaller amounts [33,34]. Shiga toxin inhibits protein synthesis in host cells [35,36]. The pathogenesis of S. dysenteriae is primarily related to its invasive phenotype, rather than Shiga toxin production [37]. However, Shiga toxin exacerbates mucosal damage via vasculitis [38], as infections with S. dysenteriae strains harboring mutations in the Shiga toxin gene in macaque monkeys lack the phenotypes of bloody stools, intestinal ischemia, and leukocyte decrease [39].

2.2. Phagosomal lysis

We overview the entry of S. flexneri in Figure 1.
After being internalized, S. flexneri escapes from the acidic environment of phagosomes. This escape is dependent on lipid rafts and involves the recruitment of Gal-3 and TECPR1. Once in cytosol, S.flexneri is captured by the autophagosome and is eventually degraded.
Multiplication of S. flexneri requires the activity of phagosomal lysis. A non-invasive strain of S. flexneri was phagocytosed; however, phagosomal lysis was not observed [40]. Invasion plasmid antigen B (IpaB), an effector secreted by the T3SS, is essential for phagosomal lysis by S. flexneri [41]. IpaB binds to CD44, a hyaluronan receptor on the cell surface [42]. CD44 localizes to the plasma membrane and induces actin foci at the entry site of S. flexneri. An Anti-CD44 antibody inhibits internalization of S. flexneri. The IpaB and CD44 complex is partitioned into detergent-resistant membranes (DRMs), and S. flexneri entry is inhibited by methyl-β-cyclodextrin (MβCD), a reagent that extracts cholesterol from membranes, suggesting that the entry of S. flexneri is dependent on lipid raftsmembrane microdomains enriched in cholesterol and sphingomyelin [43].
IpaB forms K+ ion channels in an acidic environment (pH 4), suggesting that IpaB forms pores in endosomes or lysosomes rather than in the plasma membrane [44]. Reduced activity of IpaB at pH 7.0 might be beneficial for S. flexneri. The feature of pH- and cholesterol- dependent pore formation is found in pore-forming toxins (PFTs) of Gram-positive bacteria, such as Listeriolysin O (LLO) from L. monocytogenes. L. monocytogenes is taken up through phagocytosis, then it perforates the phagosomal membrane through secretion of LLO and the phospholipases PlcA and PlcB [45,46]. LLO is a protein belonging to the cholesterol-dependent cytolysin (CDC) family [47], which is secreted as a soluble monomer that binds to cholesterol-rich lipid rafts on the phagosomal membrane, forming oligomers and maturing into transmembrane pores [48]. LLO exhibits maximum activity in an acidic environment (pH 5.5–6.0), allowing L. monocytogenes to escape the phagosome before it fuses with the lysosome [49]. Bacteria expressing LLO replicate more than bacteria expressing perfringolysin O (PFO), another CDC family PFT that lacks pH sensitivity [50,51]. Conceivably, reduced activity of LLO at pH 7.0 may protect host cells from plasma membrane damage, allowing for bacterial replication in the cytoplasm without inducing host cell death [51,52].
S. flexneri utilizes VirG for its movement by host cell actin [53,54]. VirG induces autophagy of S. flexneri by binding to ATG5, an essential protein for autophagosome formation [14,15]. IcsB secreted from S. flexneri binds to VirG to compete against ATG5 binding to VirG; therefore, wild-type S. flexneri prevents autophagy. S. flexneri ΔicsB can induce autophagy, and LC3 is recruited to S. flexneri [14] (See also Section 3-3-2). However, LC3 can be recruited to damaged membranes via membrane remnants damaged by internalized Latex beads [55] or LLOMe treatment [56]. In this case, LC3 recruitment would be independent of bacteria-secreted VirG.
Membrane remnants damaged by S. flexneri also recruit inflammasome components, including Ipaf (NLRC4) and ASC with Caspase-1; furthermore, inhibition of the autophagy pathway exacerbates proinflammatory responses, such as cytokine and reactive oxygen species (ROS) production and, eventually, necrosis [57]. In addition, The NLRC4 complex with NAIPs binds to the components of the T3SS [58,59]. Therefore, Immune reaction and autophagy events could be caused by several different pathways, and whether the effects originate from the damaged membrane of host cells or bacterial proteins, including VirG or T3SS, should be carefully assessed.

3. Molecular Detection of Membrane Damage in Mammalian Cells

We list the key factors to be recruited to damaged organelles in Table 1 following this review.
This table lists the molecules described in this review. The timing column indicates when each molecule is recruited to damaged organelles after damaging-inducing treatments, based on the published literature. Much earlier time points may be identified in future studies using improved live-cell imaging or advanced microscopy.

3.1. Galectins

3.1.1. Galectin-3

S. flexneri escapes from phagosomes and replicates in the cytosol by regulating the actin cytoskeleton of host cells [17,54]. During the entry of S. flexneri into HeLa, J774 macrophage, or CHO cells, Galectin-3 (Gal-3) is recruited to the membrane surrounding the bacterium [57,60]. Galectins are glycan-binding proteins, and Gal-3 preferentially binds to LacNAc (Galβ(1-4)GlcNAc), which is present internally in glycan residues [61,62]. Gal-3 is recruited to the phagosomal membrane, encapsulating the bacterium and a tubular structure connected to the limiting membrane of phagosomes. The glycan-binding ability of Gal-3 is required for this localization, suggesting that it binds to glycans on the internal leaflet of phagosomes rather than to lipopolysaccharide (LPS) that S. flexneri has on its cell wall [60]. Gal-3 can be recruited to membrane remnants of endosomes or lysosomes damaged by a wide variety of methods. Gal-3 is recruited to damaged phagosomes via internalized latex beads coated with transfection reagents and Salmonella-containing vacuoles (SCVs) by internalized Salmonella Typhimurium (S. Typhimurium) [55]. Early endosomes damaged by magnetic nanoparticles under a magnetic field can recruit Gal-3 [63]. Damaged lysosomes with L-leucyl-L-leucine methyl ester (LLOMe) also recruit Gal-3 [56]. As a non-bacterial damaging agent, LLOMe is often used for lysosomal membrane damage. LLOMe is a lysosome-damaging reagent that destabilizes the lysosomal lipid bilayer by forming a polymer from a dipeptide of Leucine in a Cathepsin C-dependent manner [64].
Gal-3 colocalizes with internalized S. flexneri together with ubiquitin, p62 and LC3 [57]. p62 is an autophagy adaptor that binds to polyubiquitin and bridges it to LC3[65,66]. Upon LLOMe treatment, K48- and K63-linked ubiquitin chains are conjugated to damaged lysosomes [67]. Screening of E2 ubiquitin ligases identified UBE2QL1, and both UBE2QL1 and K48- and K63-linked ubiquitin are detected 30-60 min after LLOMe treatment. Depletion of UBE2QL1 abolishes ubiquitin signals as well as the recruitment of LC3 and p97, which is required for clearance of damaged lysosomes [67,68]. Interestingly, Gal-3 recruitment to damaged lysosomes is enhanced by UBE2QL1 depletion, indicating that Gal-3 is recruited independently of ubiquitination, but Gal-3 accumulation is indirectly influenced by impaired clearance of damaged lysosomes.
Gal-3 interacts with TRIM16 to recruit ATG16L to damaged lysosomes [69]. ATG16L is a component of the autophagy machinery that is recruited to ubiquitin-positive Salmonella and ubiquitin-positive Latex-beads in a manner dependent on ubiquitin and FIP200 [55]. ATG16L determines the localization of LC3[70]. Depletion of TRIM16 inhibits ubiquitination of damaged lysosomes and partially reduces LC3 recruitment [69]. Likewise, depletion of Gal-3 partially decreases LC3 recruitment upon LLOMe treatment [71]. Thus, Gal-3 and TRIM16 contribute to LC3 recruitment and promote autophagy of damaged organelles, at least in part. However, autophagy can be triggered by several different signals, not only by Gal-3, as described in sections 3-2-3, 3-3-2 and 3-5.
Gal-3 KO mice were more susceptible to infection by Mycobacterium tuberculosis [69]. However, Gal-3 has also been reported to downregulate antibacterial autophagy [72]. Deletion of Gal-3 resulted in increased LC3 recruitment to phagosomes in L. monocytogenes-infected macrophages, which implies that Gal-3 plays a role in suppressing LC3 recruitment to phagosomes in this case [72]. Depletion of Gal-3 may inhibit the repair process of phagosomes, then LC3 recruitment could be increased. Whether Gal-3 inhibits or promotes antibacterial autophagy may depend on various conditions.

3.1.2. Galectin-8, 9

At 1 hour after infection with the Gram-negative bacteria Salmonella Typhimurium (S. Typhimurium) in HeLa cells, Gal-3, -8, and -9 were recruited to Salmonella-containing Vacuoles (SCVs) [73]. Only Gal-8 depletion increased proliferation of S. Typhimurium, suggesting that Gal-3 and Gal-9 do not play roles in inhibiting the proliferation of S. Typhimurium. Gal-8 binds similarly to LacNAc as Gal-3 [62], but binds to another autophagy adaptor, NDP-52 rather than p62 [74]. Depletion of NDP-52 increased proliferation of S. Typhimurium[73], supporting the roles of NDP-52 and Gal-8 in antibacterial activity against S. Typhimurium. After 4 hours, Gal-8-independent recruitment of NDP-52 was observed, indicating that Gal-8 is an early detector for S. Typhimurium.
Gal-9 was reported to be recruited to damaged endosomes/lysosomes by damaging agents—namely, chloroquine and siramesine—more than Gal-3 or Gal-8 [75]. Gal-9 was used for high-throughput screening of efficient lipid nanoparticle formulation [76]. In cells, Gal-9 depletion resulted in less ubiquitination of damaged lysosomes [77]. Through LLOMe treatment, Gal-9 binds to the deubiquitinating enzyme, USP9X, to inhibit deubiquitination to induce downstream effects of ubiquitination such as autophagy [77]. Depletion of Gal-9 inhibited ubiquitination of lysosomes via LLOMe treatment and inhibited LC3 lipidation [77].
Although Gal-9 has been used to detect endosomal escape, it may also affect immune system. Gal-9 is expressed in a wide variety of immune cells, and soluble Gal-9 binding to Tim-3 on the surface of CD4+ T cells suppresses T cell activity [78]. Gal-9 binding to Tim-3 downregulates coreceptors of CD4+ T cells, and upregulates p21 to inhibit HIV infection of CD4+ T cells. In contrast, Gal-9 binding to secreted PDI on the surface of CD4+ T cells enhances HIV infection to resting CD4+ T cells [79]. The studies of Gal-9 highlight important roles of Gal-9 in immune regulation.

3.2. ESCRTs

3.2.1. Overview of ESCRT Proteins

Endosomal sorting complexes required for transport (ESCRT) proteins have been identified as class E vacuolar protein sorting (Vps) proteins, including ESCRT-0, I, II, III, and the Vps4 ATPase [80,81,82,83,84,85]. Studies on ESCRT proteins in the trafficking of the epidermal growth factor receptor (EGFR), a well-known receptor tyrosine kinase [86,87,88], have deepened our understanding of the roles of the ESCRT complexes. Upon EGF binding, EGFR is ubiquitinated [89,90,91,92,93], and Hrs and STAM complexes and ESCRT-0 components bind to the ubiquitinated EGFR and transfer EGFR to ESCRT-I, II, and III, inducing inward budding of the limiting membrane to produce intraluminal vesicles (ILVs) that form multi-vesicular bodies (MVBs) [2,94,95]. Through inward budding, the phosphorylated cytosolic region of EGFR—which binds signaling complexes such as Grb2 [96,97]—is incorporated into ILVs and degraded in lysosomes together with the luminal region of EGFR [87,88]. CHMP4A and CHMP4B, which are ESCRT-III proteins, form filaments [98], and ESCRT-III mediates membrane scission to form ILVs [99,100]. ESCRT-III assembles at the bud neck and constricts the membrane with Vps4 to sever the remaining membrane [101,102]. Thus, ESCRT proteins form vesicles by pushing the membrane from the cytosol into the lumen, which is an inverse mechanism compared to coat proteins such as clathrin or COPI, which invaginate vesicles by pulling the membrane from the lumen to the cytosol. At present, ESCRT proteins are known to have many functions beyond ILV formation, and their unique membrane scission activities plays important roles in diverse cellular events [95].

3.2.2. ESCRT in Plasma Membrane Damage

When the plasma membrane was damaged by a UV laser, CHMP4B—an ESCRT-III component—was recruited to the wound before the initiation of repair [103]. The depletion of CHMP4B decreased cell survival after wounding, indicating that the ESCRT machinery functions in the wound repair of the plasma membrane [103]. ALIX, which binds to TSG101 and CHMP4B to connect Tsg101/ESCRT-I to ESCRT-III [104,105,106,107], is recruited to damaged plasma membrane [103,108]. ALIX was originally found as a protein binding to Apoptosis-linked-gene2 (ALG2), and thus was named ALG-2 interacting protein 1 or X (AIP1/ALIX) [109,110]. Overexpression of ALIX is known to induce apoptosis [111]. ALIX binds to ALG2 in a Ca2+-dependent manner, and ALG-2 is known to show oscillatory localization from cytosol to ER-exit site dependent on Ca2+ [109,110,112,113]. In steady state, ALIX localizes in endosomes dependent on lysobisphosphatidic acid (LBPA) [114,115,116]. By inducing plasma membrane damage, ALIX is recruited to the damaged area, and mutant ALIX with defective Ca2+ binding has been impaired in recruitment to the lesion of the plasma membrane, suggesting that ALIX binds to the damaged area in a Ca2+-dependent manner[103,108]. Depletion of Ca2+ also reduces CHMP4B, indicating that ALIX bypasses ESCRT-II and connects TSG101 to CHMP4B directly [104,105,106,107]. Indeed, the ESCRT-0 proteins Hrs and STAM, the ESCRT-I protein Vps37, and the ESCRT-II protein Vps25 are not detected at the damaged plasma membrane [103,108]. ALG2 is required for recruiting ALIX and CHMP4B [108], suggesting that Ca2+-dependent binding of ALG2 to the damaged area is critical for the detection of plasma membrane damage. Ubiquitin is also observed at damaged plasma membrane, but only after CHMP4B, indicating that ubiquitin conjugation is a later event than ESCRT recruitment[103]. It has been proposed that ESCRT-III and Vps4 remove damaged membrane by constricting the undamaged membrane, leading to shedding of the damaged membrane as vesicles outside the cell [103]. In recent research, ALIX is not always required for the repair of plasma membrane damage, whereas TSG101 plays an essential role [117]. ALG-2 can also bind to another protein IST1, which forms a complex with CHMP1, a component of ESCRT-III [85,118] (see Section 3-2-3). Furthermore, there are other Ca2+-dependent proteins such as IQGAP1 (see Section 3-6-2) and Annexins (see Section 3-7). Therefore, there may be several pathways for plasma membrane repair.

3.2.3. ESCRT in Lysosomal Membrane Damage

ESCRTs repair damaged lysosomal membranes as well [119,120]. The lysosome-damaging agents, LLOMe, and glycyl-L-phenylalanine 2-naphthylamide (GPN)—which is a substrate of Cathepsin C and causes osmotic lysis of lysosomes specifically [121,122,123]—have been shown to recruit the ESCRT proteins ALIX and CHMP4B, preceding the recruitment of Gal-3, ubiquitin, and LC3. The combined depletion of ALIX and TSG101 inhibited the recovery of lysosomes after the washout of damaging reagents [119,120]. These results indicate that, similarly to lesions in the plasma membrane, ALIX, TSG101, and ESCRT-III are recruited to damaged lysosomes. In addition, Ca2+ helps to recruit CHMP4B [120]. Ca2+ is required for the recruitment of ALIX to damaged lysosomes in the early phase, such as 10 minutes after lysosomal damage [71]. After 30 minutes of lysosomal damage, Gal-3 depletion decreased ALIX and CHMP4B recruitment to damaged lysosomes, as did Ca2+ chelation. Gal-3 is known to interact with ALIX [124,125], and forms a complex with ALIX and CHMP4B at 30 minutes after lysosomal damage [71]. Therefore, the initial recruitment of ALIX is mainly dependent on Ca2+; meanwhile, in later phases, ALIX can be recruited to damaged lysosomes by Gal-3 as well as by Ca2+.
Lysosomes damaged by LLOMe or silica nanoparticle, recruit IST1 and SPG20 [126]. IST1 is an ESCRT-III-like protein with a CHMP-like domain [118]. IST1 binds to ALG-2 in a Ca2+-dependent manner and forms a complex with CHMP1A and B [118], suggesting that IST1 acts as another Ca2+-dependent adaptor linking ALG-2 to ESCRT-III. Although IST1 binds to SPG20, deletion of the IST1-binding domain in SPG20 only partially reduces SPG20 recruitment to damaged lysosomes [126]. SPG20 has an SC domain that binds to the lipid loosely packed due to peroxidation. Lipid peroxidation induces SPG20 recruitment to damaged lysosomes, whereas Ca2+ release without lipid-packing defects—induced by ML-SA1, an agonist of lysosomal Ca2+ channel TRPML1— does not recruit SPG20, even though IST1 can be recruited by this condition [126]. These findings indicate that SPG20 interacts with both defective lipids and IST1. SPG20 forms a complex with ubiquitin ligase ITCH, which is recruited to damaged lysosomes. Depletion of ITCH reduces K63-linked ubiquitination in lysosomes, leading to decreases in LC3 recruitment and cell survival rates [126].

3.2.4. ESCRT in Endosomal Membrane Damage

Recruitment of ALIX, CHMP4B, and CHMP1B has been observed in damaged early endosomes, and the depletion of ALIX and TSG101 inhibits CHMP4B recruitment [127]. The rupture of early endosomes was achieved via hypertonic shock, and the reduction in membrane tension in early endosomes was correlated with the recruitment of ESCRT proteins [127]. Membrane tension was detected by a tension probe, Flipt-R, whose fluorescent lifetime decreases when membrane tension decreases [128,129]. Mercier et al. proposed that a decrease in membrane tension in early endosomes, caused by membrane damage due to hypertonic shock or by the fusion of many endocytic vesicles with endosomes upon EGF treatment, would trigger ESCRT recruitment [127].

3.2.5. ESCRT in Bacteria-Containing Vacuoles

Coxiella burnetii—a Gram-negative bacterium that proliferates in lysosomes—also caused ESCRT recruitment to lysosomes [119,120]. Salmonella-containing vacuoles (SCVs) are specific organelles in which Salmonella proliferate without escaping into the cytosol. The depletion of CHMP3, an ESCRT-III protein, induced the enlargement of SCVs and significantly increased the proliferation of Salmonella Typhimurium in the cytosol [130]. ESCRT proteins likely play roles in controlling the shape and size of SCVs to inhibit bacterial proliferation.

3.3. Sphingomyelin

3.3.1. Sphingomyelin Exposure in Phagosomal Escape

Sphingomyelin is an important component of lipid rafts. Lipid rafts are membrane microdomains that are initially characterized by DRMs and are sensitive to MβCD, which eliminates cholesterol from the liquid-disordered phase of the membrane [131,132]. Sphingomyelin is synthesized from ceramide in the lumen of the Golgi apparatus [133] and is transported to the plasma membrane, endosomes, and lysosomes [134,135,136,137]. Sphingomyelin resides in the luminal leaflet of organelles or the outer leaflet of the plasma membrane [134]. Ellison et al. reported that the bacterial membrane damage caused by S. Typhimurium, S. flexneri, L. monocytogenes, and Streptococcus pyogenes exposes sphingomyelin to the cytosolic side [138]. The cytosolic presence of sphingomyelin is detected by the C-terminal region of Lysenin, a sphingomyelin-binding proteinaceous toxin isolated from the earthworm Eisenia foetida [139]. Lysenin recruitment to S. Typhimurium and S. flexneri preceded that of Gal-8. Through electron microscopy, damaged membranes positive only for Lysenin were shown to have 100-200 nm gaps, whereas those double-positive for Lysenin and Gal-8 had large gaps and no longer enclose S. Typhimurium, suggesting that sphingomyelin exposure to the cytosol is an earlier event than Galectin recruitment [138].

3.3.2. Sphingomyelin Receptor in Membrane Damage

Sphingomyelin can also be detected by equinatoxin II (EqtII), a PFT isolated from Actinia equina [140], and EqtII-GFP detects organelles after permeabilization [141,142]. Interestingly, Lysenin and EqtII do not colocalize completely [142]. In the presence of glycolipids that are miscible with sphingomyelin, the binding of Lysenin to SM was decreased [143], indicating that Lysenin binds to highly clustered sphingomyelin but not to sphingomyelin mixed with glycolipids. Lysenin stained large puncta in the plasma membrane as well as sphingomyelin in late endosomes, but not in the Golgi apparatus, early endosomes, or recycling endosomes [142]. In contrast, EqtII detects sphingomyelin in smaller puncta in the plasma membrane as well as in late endosomes and recycling endosomes [142]. EqtII can bind to sphingomyelin dispersed in glycolipids, thus EqtII can detect more sphingomyelin in diverse organelles.
TECPR1 was revealed as an endogenous receptor for sphingomyelin exposed to the cytosol [144]. TECPR1 is known to bind to ATG5 [145], and is localized in S. flexneri ΔicsB-positive membrane. IcsB, a protein encoded by the virulence plasmid in S. flexneri[146], binds to VirG[14]. IcsB usually inhibits the interaction between VirG and ATG5 to inhibit autophagy of S.flexneri, and depletion of IcsB in S. flexneri inhibited the escape of S. flexneri from the autophagosome. As such, S. flexneri ΔicsB was trapped in the autophagosome [14,146]. TECPR1 depletion inhibited the recruitment of LC3 to the S. flexneri ΔicsB-positive membrane, and replication of S. flexneri ΔicsB increased [145]. In addition, TECPR1 localized to damaged membranes caused by S. typhimurium and L. monocytogenes, as well as those damaged by osmotic shock and LLOMe [144]. TECPR1 promotes LC3 conjugation to sphingomyelin-containing liposomes with ATG5 and ATG12, demonstrating that sphingomyelin indeed participates in autophagy pathway involved in organelle damage [144].

3.3.3. Sphingomyelin Pathway in Membrane Damage

Sphingomyelin is exposed by Ca2+-activated scramblase, rather than through simple exposure upon membrane damage [147]. EqtSM—an SM binding protein expressed in cytosol—is recruited to damaged organelles upon treatment with LLOMe or Streptolysin O (SLO), a PFT from S. pyogenes, and the removal of Ca2+ by EGTA or BAPTA-AM reduced EqtSM recruitment to damaged organelles [147]. The depletion of TMEM16F—a calcium-activated scramblase localized in the plasma membrane—abolished EqtSM recruitment to SLO- and ionomycin-induced damaged membranes but not to lysosomes damaged by LLOMe, suggesting that TMEM16F plays a role in the plasma membrane but not in lysosomes. As BAPTA-AM also inhibited the recruitment of EqtSM to lysosomes damaged by LLOMe, there is likely another scramblase in lysosomes. The depletion of ESCRT proteins, including CHMP3, ALIX, and TSG101, did not inhibit EqtSM recruitment, suggesting that sphingomyelin exposure is independent of ESCRT proteins [147]. The cell survival after LLOMe treatment was reduced due to the depletion of either sphingomyelin synthase or ALIX and TSG101, indicating that the sphingomyelin pathway contributes to cell survival independent of the ESCRT pathway [147].

3.4. Stress Granules

Lysosomal damage induces stress granule formation. Various stress granule components were found to be associated with lysosomes via lysosomal IP after LLOMe treatment [148]. G3BP1 is a core protein to drive stress granule assembly by interacting with cytosolic RNAs by liquid–liquid phase separation [149,150]. Interestingly, stress granule formation occurs in a different location than lysosomes. After their formation, stress granules associate with damaged lysosomes [148]. The stress granule protein, NUFIP2, supports the inactivation of mTOR upon lysosomal damage to induce autophagy. Depletion of NUFIP2 keeps mTOR active, resulting in maintained autophagy pathway inactive [148]. G3BP1 forms condensates in damaged artificial vesicles with poly(A)-RNA at low pH, implicating the mixing of lower osmolarity and low pH solution with RNA leading to condensation of G3BP1[151]. Depletion of G3BP1 and 2 increased replication of M. tuberculosis, suggesting that stress granule plugs restrict bacterial proliferation in host cells [151]. Stress granule formation is triggered by various stresses, leading to phosphorylation of eIF2α and inhibiting global translation, thus resulting in the accumulation of untranslated mRNAs [152]. A search for proteins binding to eIF2α under LLOMe treatment identified PKR and its activator PACT as upstream kinases upon lysosomal damage to eIF2α [153]. ALIX binds to PKR and PACT under LLOMe treatment, and depletion of Ca2+ or ALIX inhibits phosphorylation of eIF2α, suggesting that Ca2+ leakage and recruitment of ALIX/ALG2 lead to phosphorylation of eIF2α for stress granule formation. The stress granule protein, NUFIP2, binds to Gal-8; however, depletion of Gal-8 does not inhibit stress granule formation [148]. On the other hand, depletion of Gal-3 increases stress granule formation, indicating that the Gal-3 pathway contributes to the repair of lysosomal damage [153]. Stress granule formation can be induced by various pathogenic lysosomal damage, including M. tuberculosis, Adenovirus infection, SARS-Cov-2 ORF3a protein, malarial pigment hemozoin, silica crystals, and Tau aggregates [148,153], indicating the importance of stress granule formation in attenuating the effects of lysosomal damage.

3.5. Rabaptin-5 in Endosomal Damage

Rabaptin-5 is a protein that binds to the small GTPases Rab5 and Rab4 and localizes to early and recycling endosomes [154,155,156,157]. Early and recycling endosomes are damaged by Chloroquine and Monensin [158]. Chloroquine accumulates in acidic compartments, where it absorbs protons and induces swelling of endosomes or lysosomes through osmotic water influx. Monensin is an ionophore that perturbs the exchange of Na+ and H+, thereby preventing acidification, primarily in recycling endosomes and the Golgi apparatus. Chloroquine treatment induces recruitment of Rabaptin-5, Gal-3, Gal-8, ubiquitin and LC3 to endosomes [158]. Rabaptin-5 binds to the autophagy initiators FIP200 and ATG16L, and depletion of Rabaptin-5 inhibited LC3 recruitment to early or recycling endosomes [158]. Depletion of Rabaptin-5 increased the survival of Salmonellae internalized in HeLa and HEK293A cells, suggesting Rabaptin-5 play a role in restriction of Salmonellae survival.

3.6. Membrane Contact Sites in Membrane Repair

3.6.1. Phosphatidylinositol-4 Phosphate (PI4P) in Lysosomal Damage

Phosphatidylinositol-4 phosphate (PI4P) localizes in the Golgi, endosomes/lysosomes and the plasma membrane [159]. PI4P serves as a binding site of lipid transfer proteins, including Oxysterol-binding protein (OSBP) and OSBP-related proteins (ORPs) [159,160,161]. ORPs often localize in membrane contact sites in a PI4P-dependent manner at post-Golgi organelles where ORPs mediate the transfer of sterols or phosphatidylserine (PS) between post-Golgi organelles and the ER membrane. PI4P can be visualized using PI4P-binding probes such as the PD domain of OSBP (OSBP-PH) and SidM(P4M) [162,163]. Localization of OSBP-PH to the Golgi depends on both PI4P and Arf1, a small GTP-binding protein essential for vesicle budding at the Golgi[164]. Treatment of Brefeldin A (BFA), which inhibits the guanine nucleotide exchange factor required for Arf1 activation [165,166,167], disrupts Golgi localization of OSBP-PH [162]. This suggests that Golgi targeting of OSBP-PH requires not only PI4P but also Arf1. In contrast, SidM—a protein secreted by Legionella pneumophila—is solely dependent on PI4P, as SidM remains at the Golgi even after BFA treatment [163].
Proteomic and lipidomic analyses of damaged lysosomes revealed that PI4P is generated upon lysosomal injury [168,169]. Consistently, SidM is recruited to damaged lysosomes 5-10 min after LLOMe [168]. OSBP-PH is also recruited to damaged lysosomes[169]. Notably, OSBP-PH recruitment to damaged lysosomes after LLOMe treatment is independent of Arf proteins because BFA treatment does not inhibit the recruitment of OSBP-PH [169]. PI4P production at damaged lysosomes requires PI4KIIA, since PI4KIIA depletion abolishes OSBP-PH recruitment to damaged lysosomes [169]. PI4KIIA itself is recruited to damaged lysosomes about 10 min after LLOMe treatment [169]. Importantly, activation of the lysosomal Ca2+ channel TRPML1 by its agonist ML-SA1 is sufficient to trigger PI4KIIA recruitment, suggesting that Ca2+ leakage is a key signal for recruitment of PI4KIIA to damaged lysosomes [169]. Interestingly, PI4KIIA recruitment is independent of ESCRT machinery. Depletion of ALIX and TSG101 does not affect PI4P production in damaged lysosomes, and conversely, depletion of PI4KIIA does not impair the recruitment of ALIX, CHMP4B, and Gal-3 to damaged lysosomes [168].
Within 10 min after LLOMe treatment, ORP1L, ORP9, 10, 11, and OSBP are recruited to damaged lysosomes [168,169]. ORP9, OSBP, and ORP1L contain an FFAT motif that binds to VAP-A and VAP-B localized in the ER and form membrane contact sites [159,161]. LLOMe treatment extensively increased ER–lysosome membrane contact sites [168,169]. Depletion of VAP-A/B attenuates the formation of ER–lysosome membrane contact sites [168]. Similarly, ORP9, 10, 11, and OSBP quadruple knockout (QKO) diminished VAP-A clustering around damaged lysosomes [169], suggesting that VAP-A/B and ORP form membrane contact sites in lysosomes upon lysosomal damage. ORP9 and 11 transfer phosphatidylserine (PS) to PI4P liposomes in vitro, and LLOMe treatment increased PS in lysosomes [169]. PS accumulation is rescued by any of two ORPs, ORP9/10, 9/11, or 10/11, and reduced Gal-3 recruitment to damaged lysosomes, implicating that PS transfer to damaged lysosomes plays a role in the repair of lysosomes.
Cholesterol also plays a role in repairing damaged lysosomes. OSBP transfers cholesterol, and depletion of OSBP decreased the survival of cells after LLOMe treatment. Accumulation of cholesterol in lysosomes decreases Gal-3 recruitment to damaged lysosomes, indicating that the presence of cholesterol protects lysosomes from damage as well as PS [168].

3.6.2. ATG2-ATG9 in Membrane Damage

ATG2, originally discovered as a required component for autophagy in yeast [15,16,170,171], possesses a cavity to bind to lipids and, thus, has lipid transfer activity [172,173,174]. Similarly, the human homologs ATG2A and ATG2B have PS transfer activity [174,175]. ATG2A localizes in lysosomes after LLOMe treatment [169]. Expression of mutant ATG2A that lost lipid transfer activity of PS to PI4P-positive liposomes in vitro maintains Gal-3 puncta longer than WT ATG2A expression, suggesting that PS transfer of ATG2A to lysosomes helps to repair damaged lysosomes [169].
ATG9, another essential protein for autophagy [176,177], helps to repair the damaged plasma membrane [117] and lysosomes [178]. ATG9 is a multi-membrane spanning protein and cycles between TGN and late and recycling endosomes [179,180]. When the autophagy pathway is activated, ATG9 relocalizes to precursors of the autophagosome [180,181]. ATG9 forms a homotrimer complex [182] and has lipid scramblase activity [183]. ATG9 binds to ATG2 and is thought to supply phospholipids to the autophagosome for autophagosome membrane expansion [183,184]. ATG9 binds to many of the vesicle budding machineries. ATG9 binds to clathrin adaptors, AP-2 [185,186], AP-4 [187,188], AP-1 [189], and AP-3 [178], as well as retromer complex [185]. When AP-2 or AP-4 are depleted, ATG9 remains in the recycling endosomes or TGN even under starvation conditions, and the autophagy pathway is inhibited, suggesting proper transport of ATG9 is required for autophagosome formation [185,186,187].
When the plasma membrane was damaged by digitonin, saponin, SLO, or the endogenous pore-forming protein Gasdermin-D, plasma membrane permeabilization was exacerbated in ATG9-depleted cells indicating that ATG9 protects plasma membrane from damage [117]. ATG9 is recruited to the injured plasma membrane in a manner dependent on IQGAP1, which responds to Ca2+, and depletion of Ca2+ outside of cells inhibits ATG9 recruitment to the injured plasma membrane [117]. IQGAP1 interacts with ATG9 and CHMP2A, ESCRT-III protein, and double knockout of ATG9 and CHMP2B does not exacerbate PM permeabilization compared to single knockout of ATG9 or CHMP2B, which indicates that ATG9 and CHMP2B function in the same pathway for plasma membrane repair.
ATG9 has also been reported to play a role in lysosomal repair as well [178]. In LLOMe-treated cells, ATG9 is recruited to damaged lysosomes [178]. Depletion of cellular Ca2+ inhibited ATG9 recruitment, indicating that Ca2+ promotes ATG9 recruitment [178]. ATG9 vesicles contain ARFIP2/Arfaptin 2, an Arf-GTP binding protein [164], and bind to PI4P and PI4PK2A [178,190]. Depletion of ARFIP2 increases ATG9, PI4KIIA, and PI4P localization in damaged lysosomes, and lysosomes are repaired faster [178]. ARFIP2 forms a complex with AP-3 and, when AP-3 is depleted, ATG9 increases in damaged lysosomes, implicating that AP-3 helps to retrieve ATG9 from the endolysosomal compartment to the Golgi and attenuates lysosomal repair [178]. ARFIP2 binds to ORP9 and interferes with the PS lipid transfer activity of ORP9/11 in vitro [178]. Depletion of ARFIP2 restricts M. tuberculosis and Salmonella infection, demonstrating that ARFIP2 inhibits lysosomal repair under normal conditions, while depletion of ARFIP2 promotes lysosomal repair [178]. These findings indicate that the transfer of PS and cholesterol at the expense to PI4P contributes to membrane repair, mediated by ORP9-11, OSBP, or ATG2-ATG9 complex. In contrast, clathrin adaptors and ARFIP2 restrict the repair process by regulating ATG9 trafficking.

3.7. Annexins

3.7.1. Annexins in Plasma Membrane Repair

Annexins are a family of Ca2+-dependent phospholipid-binding proteins [191,192]. Annexins bind to negatively charged phospholipids, including PS, PI, and PI(4,5) P2, in a manner dependent on Ca2+ [193,194,195,196,197,198,199]. Among them, PS is most characterized and largely used for Ca2+-dependent binding of Annexins to lipids. In addition, Annexin A2 binds to cholesterol independent of Ca2+[200].
Annexins A1 and A2 associate with Dysferlin, whose mutation causes dysferlinopathy [201]. Dysferlin-deficient myotubules lose the ability to reseal the damaged plasma membrane [201]. Lack of Annexin A2 impairs repair of injury in a Cholesterol-dependent manner in muscle cells [202]. Annexins also affect plasma membrane repair in non-muscle cells. As shown via the scraping procedure in HeLa and kidney epithelial cells, Annexin A1 deficient for Ca2+ binding loses the ability to reseal plasma membrane injury [203]. Annexins A4, 5, 6, and 7 are reportedly also involved in plasma membrane repair [204,205,206].
Annexins A1, 2, and 6 translocate to the wound area in a wave-like manner and form a ring-like structure around the wound within 2-30 sec [207]. Annexins A1, 2, 5, and 6 localize in the repair cap as well after injury in 50 sec, dependent on Ca2+ and actin [208]. Annexin A5 forms a trimer to form a 2D array via self-assembly on PS-exposed membranes [205]. An Annexin A5 mutant that disrupt its trimer structure inhibited membrane repair; however, membrane binding was not inhibited [205], suggesting that self-assembly of Annexin A5 is required for membrane repair but not membrane binding.
Annexins A4 and A6 can regulate membrane curvature [204]. Annexin A4 binds to the edge of the membrane and rolls up the membrane. In contrast, Annexin A6 can constrict the planar membrane. As determined via live imaging, Annexin A6 was recruited to the damaged area before Annexin A4 and localized as a ring-like structure around the wound. It has been proposed that Annexin A6 binds to the edge and Annexin A4 rolls up the hole of the edge, following which Annexin A6 constricts to close the membrane. This process completed within 15 sec in a mammary carcinoma cell line, MCF7 cells [204].
Annexin A7 also plays a role in plasma membrane repair [206]. Annexin A7 binds to ALG2 and recruits ALG2 to the plasma membrane wound within 100 sec. ALG2 is accumulated at the edge of the membrane in an Annexin A7-dependent manner. A mutant of Annexin A7 that cannot be recruited to the lesion of the plasma membrane inhibited ALG2 recruitment. Interestingly, CHMP4B localized at the repair site, and shed vesicles were observed from the plasma membrane within 2-15 min. The ESCRT complex induces shedding of the plasma membrane as microvesicles/ectosomes for membrane repair. The findings of these studies indicate that Annexins function as first-line machinery for plasma membrane damage detection and repair. ESCRT-mediated membrane repair follows the repair initiated by Annexin A7.

3.7.2. Annexins in Endolysosomal Repair

Annexin A2 is recruited to damaged multi-vesicular bodies (MVBs) as well as lysosomes in dendritic cells (DCs) [209]. Ultra-high molecular weight polyethylene is released from joint replacement as wear debris, and induces inflammation when endocytosed by macrophages and DCs [210]. Wear debris comprising nanometer- to micrometer-sized particles are incorporated into MVBs and release lysosomal enzymes such as Cathepsin S and B [209,210], suggesting the internalization of wear debris damages MVBs and lysosomes. Depletion of Annexin A2 results in increases in the release of Cathepsin B and IL-1b [209]; therefore, Annexin A2 would repair damaged MVBs and lysosomes.
Annexins A1 and 2 play roles in lysosomal repair after LLOMe treatment in human osteosarcoma U2Os cells [211]. Although many Annexins are recruited to damaged lysosomes, only Annexins A1 and 2 have activity for lysosomal repair. Interestingly, Annexin A1/2 depletion slows down lysosomal repair more strongly than ALIX/TSG101 depletion, suggesting that the Annexin A1/2 repair pathway is independent of ESCRTs, in contrast to plasma membrane repair (see section 3-7-1). Annexins A1 and 2 are preferably recruited to damaged lysosomes that sustain larger injuries, sufficient to release 10-KDa dextran, whereas ESCRTs can be recruited to even to lysosomes with smaller injuries.
Annexin A7 has also been reported to be important for lysosomal repair in MCF7 and HeLa cells [212]. Depletion of Annexin A7 exacerbates Gal-3 recruitment to damaged lysosomes by LLOMe. Although Annexin A7 is known to bind to ALG2 [206], CHMP4B recruitment to damaged lysosomes is not affected in Annexin A7-depleted cells [212]. In addition, Annexin A7 depletion does not affect lysophagy or alter lipid composition, which is regulated by the PI4P pathway [168,169]. Thus, Annexin A7 facilitates lysosomal repair through other mechanisms.
We also note that the timing of Annexin recruitment to damaged lysosomes is slower than to damaged plasma membrane (Table 1). Together, these findings suggest that Annexin contribute to lysosomal repair via mechanisms distinct from those operating during plasma membrane repair.

3.7.3. Annexins for Secretory Lysosome/MVB Fusion to the Plasma Membrane

Upon plasma membrane damage, it is proposed that secretory lysosomes fuse with the damaged area of the plasma membrane in a Ca2+-dependent manner to repair plasma membrane damage [213]. Annexins may facilitate such fusion for plasma membrane repair.
Annexin A2 has endosome fusion activity [214], and plays a role in exocytosis in chromaffin cells [215]. In Annexin A2-deficient chromaffin cells, exocytosis upon nicotine stimulation is inhibited [215]. Annexin A2 is translocated to the plasma membrane after exocytosis, in a manner dependent on lipid rafts. Therefore, it is proposed that Annexin A2 forms a lipid raft in the plasma membrane in a Ca2+-dependent manner and mediates the fusion of secretory granules with the plasma membrane[215].
Exosomes are endosome-derived extracellular vesicles, which are secreted by a wide variety of cells, including cancer cells and platelets [216]. When multivesicular bodies (MVBs) fuse with the plasma membrane, their internal luminal vesicles (ILVs) are released as exosomes [217,218,219]. The tetraspanin protein CD63 localizes in ILVs of MVBs and is secreted on exosomes [220]. Exosomal release can be induced by Ca2+ in HCT116, a human colon cancer cell line, and plasma membrane damage by streptolysin O (SLO) also stimulates exosome secretion [221].
Annexins A2 and 6 have been identified as proteins recruited to CD63-positive MVBs in a Ca2+ dependent manner [221]. Although the role of Annexin A2 has not been examined, depletion of Annexin A6 inhibited MVB fusion and release of exosomes stimulated by Ca2+ ionophore [221]. Moreover, an antibody against Annexin A6 prevents SLO-stimulated exosome release, indicating that Annexin A6 mediates the fusion of MVBs to the damaged plasma membrane.
These findings suggest a model in which CD63-positive MVBs fuse with the damaged plasma membrane to facilitate plasma membrane repair, releasing exosomes. However, whether this mechanism represents a ubiquitous cellular response or is restricted to certain secretory or cancer cells remains to be determined (see Future Perspectives).

4. Future Perspective

Studies on bacteria have significantly advanced our understanding of how cells detect organelle membrane damage. Some molecules are shared between the detection of plasma membrane and lysosomal membrane damage, whereas others are specific to each organelle. The key molecules are summarized in Table 1, and Figure 2 illustrates their association with each organelle.
We summarize the timing of recruitment of each molecule, as reported in the studies discussed in this review in Table 1. Early recruitment events are largely dependent on calcium release, which triggers the recruitment of ALG2/ALIX with the ESCRT complex, ATG9 vesicles, and Annexins. These events occur at both the plasma membrane and damaged lysosomes, although not all molecules are universally present (Table 1 and Figure 2). Additional mechanisms for membrane damage detection include the exposure of sphingomyelin and glycans to the cytosol, which recruits TECPR1 and Galectins, thereby initiates autophagy. Stress granule formation appears to occur specifically upon lysosomal damage, resulting from the mixing of acidic, high-osmolarity lysosomal contents with the cytosol. Moreover, Rabaptin-5-mediated signaling from early or recycling endosomes to the autophagy pathway also seems to be unique to early/recycling endosomes, as Rabaptin-5 binds to Rab4 and Rab5, which are typically specific to early/recycling endosomes. Future studies should aim to reveal how specific each event is to each organelle.
This figure illustrates the key factors involved in membrane damage detection in different organelles, as described in this review. Various artificial methods used to induce organelle damage are also indicated. Abbreviations not used in the main text: NP, nanoparticle; MNP, magnetic nanoparticle; MF, magnetic field; UHMWPE, Ultra-high molecular weight polyethylene.
Although secretory MVBs were not the focus of this review, the discussion of membrane damage detection and repair naturally converges on the question of how secretory MVBs contribute to plasma membrane repair. The hypothesis that secretory MVBs or lysosomes fuse with the plasma membrane to repair damage is attractive, but several issues remain unresolved. First, it is unclear whether this fusion occurs universally or only in specific cell types. CD63-positive MVBs are positive for Rab27, a secretory granule marker [222], and fusion with the plasma membrane can be stimulated by Ca2+ or even by histamine in HeLa cells, suggesting even non-classical secretory cells may harbor secretory granule-like MVBs [223]. In contrast, CD63 release has been reported to occur independently of Rab27 in HEK293 cells [224]. Thus, there may be cell types that lack “secretory” MVBs or lysosomes to fuse with the plasma membrane. Second, even if cells have secretory MVBs or lysosomes to fuse with the plasma membrane, these cells may have several different MVBs. It is known that there are MVBs that are only EGF-positive, only CD63-positive, and both-positive [225,226]. CD63-positive MVBs are considered secretory MVBs that release exosomes via plasma membrane fusion [221]. Our analyses of endosomal sorting by ArfGAP proteins, which hydrolyze Arf-GTP and regulate cargo sorting [227,228,229], revealed that EGF trafficking to lysosomes is regulated by ArfGAP3 [230], whereas CD63 to MVBs is regulated by ADAP1 or ARAP1, other ArfGAP proteins [226]. Therefore, even if cells have secretory MVBs, these may be distinct from normal “degradable” MVBs, which are typically EGF-positive. Third, it remains unclear whether secretory MVBs are completely different from degradable MVBs or not. EGF-positive MVBs can fuse with the plasma membrane when the lysosomal pathway is inhibited [231]. In addition, Rab7-positive MVBs/lysosomes, which are usually destined for degradation, can mature into Rab27-positive secretory MVBs through the formation of membrane contact sites [232]. Thus, it is possible that secretory MVBs are derived from normal degradable MVBs. Determining which cell type possess secretory MVBs, which MVBs are responsible for repairing plasma membrane damage, and how secretory MVBs are formed or diverge from degradable MVBs should be the focus of future work.
Finally, membrane damage responses are increasingly recognized as integral components of broader cellular stress responses, with implications extending to exosome biology, autophagy, inflammation, and the development of Drug Delivery Systems (DDS). Continued discoveries are expected in this field to expand the list of key factors involved, thereby contributing to a broad range of biomedical research.

Author Contributions

Conceptualization and writing, Y.S.; figures and editing, N.S.

Funding

This work was supported by a SPERC grant of Iwate University, by The Watanabe Foundation, Grant Number 280214, by Yamada Science Foundation, Grant Number 1078 and by JSPS KAKENHI Grant Number 23K05676 to YS.

References

  1. Dong, J.; Tong, W.; Liu, M.; Liu, M.; Liu, J.; Jin, X.; Chen, J.; Jia, H.; Gao, M.; Wei, M.; et al. Endosomal traffic disorders: a driving force behind neurodegenerative diseases. Transl Neurodegener 2024, 13, 66. [Google Scholar] [CrossRef]
  2. Raiborg, C.; Rusten, T.E.; Stenmark, H. Protein sorting into multivesicular endosomes. Curr Opin Cell Biol 2003, 15, 446–455. [Google Scholar] [CrossRef]
  3. Huotari, J.; Helenius, A. Endosome maturation. EMBO J 2011, 30, 3481–3500. [Google Scholar] [CrossRef]
  4. Scott, C.C.; Vacca, F.; Gruenberg, J. Endosome maturation, transport and functions. Semin Cell Dev Biol 2014, 31, 2–10. [Google Scholar] [CrossRef] [PubMed]
  5. Murphy, R.F.; Powers, S.; Cantor, C.R. Endosome pH measured in single cells by dual fluorescence flow cytometry: rapid acidification of insulin to pH 6. J Cell Biol 1984, 98, 1757–1762. [Google Scholar] [CrossRef] [PubMed]
  6. Kielian, M.C.; Marsh, M.; Helenius, A. Kinetics of endosome acidification detected by mutant and wild-type Semliki Forest virus. EMBO J 1986, 5, 3103–3109. [Google Scholar] [CrossRef] [PubMed]
  7. Yamashiro, D.J.; Maxfield, F.R. Acidification of morphologically distinct endosomes in mutant and wild-type Chinese hamster ovary cells. J Cell Biol 1987, 105, 2723–2733. [Google Scholar] [CrossRef]
  8. Dunn, K.W.; Maxfield, F.R. Delivery of ligands from sorting endosomes to late endosomes occurs by maturation of sorting endosomes. J Cell Biol 1992, 117, 301–310. [Google Scholar] [CrossRef]
  9. Mukherjee, S.; Soe, T.T.; Maxfield, F.R. Endocytic sorting of lipid analogues differing solely in the chemistry of their hydrophobic tails. J Cell Biol 1999, 144, 1271–1284. [Google Scholar] [CrossRef]
  10. Sandvig, K.; Ryd, M.; Garred, O.; Schweda, E.; Holm, P.K.; van Deurs, B. Retrograde transport from the Golgi complex to the ER of both Shiga toxin and the nontoxic Shiga B-fragment is regulated by butyric acid and cAMP. J Cell Biol 1994, 126, 53–64. [Google Scholar] [CrossRef]
  11. Neel, E.; Chiritoiu-Butnaru, M.; Fargues, W.; Denus, M.; Colladant, M.; Filaquier, A.; Stewart, S.E.; Lehmann, S.; Zurzolo, C.; Rubinsztein, D.C.; et al. The endolysosomal system in conventional and unconventional protein secretion. J Cell Biol 2024, 223. [Google Scholar] [CrossRef]
  12. Gao, Y.; Zheng, X.; Chang, B.; Lin, Y.; Huang, X.; Wang, W.; Ding, S.; Zhan, W.; Wang, S.; Xiao, B.; et al. Intercellular transfer of activated STING triggered by RAB22A-mediated non-canonical autophagy promotes antitumor immunity. Cell Res. 2022, 32, 1086–1104. [Google Scholar] [CrossRef] [PubMed]
  13. van der Beek, J.; de Heus, C.; Sanza, P.; Liv, N.; Klumperman, J. Loss of the HOPS complex disrupts early-to-late endosome transition, impairs endosomal recycling and induces accumulation of amphisomes. Mol Biol Cell 2024, 35, ar40. [Google Scholar] [CrossRef]
  14. Ogawa, M.; Yoshimori, T.; Suzuki, T.; Sagara, H.; Mizushima, N.; Sasakawa, C. Escape of intracellular Shigella from autophagy. Science 2005, 307, 727–731. [Google Scholar] [CrossRef] [PubMed]
  15. Mizushima, N.; Yoshimori, T.; Ohsumi, Y. The role of Atg proteins in autophagosome formation. Annu Rev Cell Dev Biol 2011, 27, 107–132. [Google Scholar] [CrossRef]
  16. Ohsumi, Y. Historical landmarks of autophagy research. Cell Res. 2014, 24, 9–23. [Google Scholar] [CrossRef]
  17. Schroeder, G.N.; Hilbi, H. Molecular pathogenesis of Shigella spp.: controlling host cell signaling, invasion, and death by type III secretion. Clin. Microbiol. Rev. 2008, 21, 134–156. [Google Scholar] [CrossRef]
  18. Coburn, B.; Sekirov, I.; Finlay, B.B. Type III secretion systems and disease. Clin. Microbiol. Rev. 2007, 20, 535–549. [Google Scholar] [CrossRef]
  19. Okui, N. Dr. Kiyoshi Shiga (1871-1957): Outstanding Bacteriologist Who Discovered Dysentery Bacillus and Contributed Immensely to Public Health in Japan. Cureus 2024, 16, e71881. [Google Scholar] [CrossRef] [PubMed]
  20. Wassef, J.S.; Keren, D.F.; Mailloux, J.L. Role of M cells in initial antigen uptake and in ulcer formation in the rabbit intestinal loop model of shigellosis. Infect Immun 1989, 57, 858–863. [Google Scholar] [CrossRef]
  21. Sansonetti, P.J.; Arondel, J.; Cantey, J.R.; Prevost, M.C.; Huerre, M. Infection of rabbit Peyer's patches by Shigella flexneri: effect of adhesive or invasive bacterial phenotypes on follicle-associated epithelium. Infect Immun 1996, 64, 2752–2764. [Google Scholar] [CrossRef]
  22. Zychlinsky, A.; Prevost, M.C.; Sansonetti, P.J. Shigella flexneri induces apoptosis in infected macrophages. Nature 1992, 358, 167–169. [Google Scholar] [CrossRef]
  23. Gerber, D.F.; Watkins, H.M. Growth of shigellae in monolayer tissue cultures. J Bacteriol 1961, 82, 815–822. [Google Scholar] [CrossRef]
  24. Hale, T.L.; Morris, R.E.; Bonventre, P.F. Shigella infection of henle intestinal epithelial cells: role of the host cell. Infect Immun 1979, 24, 887–894. [Google Scholar] [CrossRef]
  25. Oaks, E.V.; Wingfield, M.E.; Formal, S.B. Plaque formation by virulent Shigella flexneri. Infect Immun 1985, 48, 124–129. [Google Scholar] [CrossRef]
  26. Bernardini, M.L.; Mounier, J.; d'Hauteville, H.; Coquis-Rondon, M.; Sansonetti, P.J. Identification of icsA, a plasmid locus of Shigella flexneri that governs bacterial intra- and intercellular spread through interaction with F-actin. Proc Natl Acad Sci U S A 1989, 86, 3867–3871. [Google Scholar] [CrossRef] [PubMed]
  27. Makino, S.; Sasakawa, C.; Kamata, K.; Kurata, T.; Yoshikawa, M. A genetic determinant required for continuous reinfection of adjacent cells on large plasmid in S. flexneri 2a. Cell 1986, 46, 551–555. [Google Scholar] [CrossRef]
  28. Maurelli, A.T.; Baudry, B.; d'Hauteville, H.; Hale, T.L.; Sansonetti, P.J. Cloning of plasmid DNA sequences involved in invasion of HeLa cells by Shigella flexneri. Infect Immun 1985, 49, 164–171. [Google Scholar] [CrossRef]
  29. Kato, J.; Ito, K.; Nakamura, A.; Watanabe, H. Cloning of regions required for contact hemolysis and entry into LLC-MK2 cells from Shigella sonnei form I plasmid: virF is a positive regulator gene for these phenotypes. Infect Immun 1989, 57, 1391–1398. [Google Scholar] [CrossRef] [PubMed]
  30. Sasakawa, C.; Kamata, K.; Sakai, T.; Makino, S.; Yamada, M.; Okada, N.; Yoshikawa, M. Virulence-associated genetic regions comprising 31 kilobases of the 230-kilobase plasmid in Shigella flexneri 2a. J Bacteriol 1988, 170, 2480–2484. [Google Scholar] [CrossRef] [PubMed]
  31. Bajunaid, W.; Haidar-Ahmad, N.; Kottarampatel, A.H.; Ourida Manigat, F.; Silue, N.; Tchagang, C.F.; Tomaro, K.; Campbell-Valois, F.X. The T3SS of Shigella: Expression, Structure, Function, and Role in Vacuole Escape. Microorganisms 2020, 8. [Google Scholar] [CrossRef]
  32. Dubos, R.J.; Geiger, J.W. Preparation and properties of Shiga toxin and toxoid. J Exp Med 1946, 84, 143–156. [Google Scholar] [CrossRef] [PubMed]
  33. Keusch, G.T.; Jacewicz, M. The pathogenesis of Shigella diarrhea. VI. Toxin and antitoxin in Shigella flexneri and Shigella sonnei infections in humans. J Infect Dis 1977, 135, 552–556. [Google Scholar] [CrossRef] [PubMed]
  34. Cantey, J.R. Infectious diarrhea. Pathogenesis and risk factors. Am. J. Med. 1985, 78, 65–75. [Google Scholar] [CrossRef] [PubMed]
  35. O'Brien, A.D.; Thompson, M.R.; Gemski, P.; Doctor, B.P.; Formal, S.B. Biological properties of Shigella flexneri 2A toxin and its serological relationship to Shigella dysenteriae 1 toxin. Infect Immun 1977, 15, 796–798. [Google Scholar] [CrossRef]
  36. Hale, T.L.; Formal, S.B. Protein synthesis in HeLa or Henle 407 cells infected with Shigella dysenteriae 1, Shigella flexneri 2a, or Salmonella typhimurium W118. Infect Immun 1981, 32, 137–144. [Google Scholar] [CrossRef]
  37. Gemski, P.; Takeuchi, A.; Washington, O.; Formal, S.B. Shigellosis due to Shigella dysenteriae. 1. Relative importance of mucosal invasion versus toxin production in pathogenesis. J Infect Dis 1972, 126, 523–530. [Google Scholar] [CrossRef]
  38. O'Loughlin, E.V.; Robins-Browne, R.M. Effect of Shiga toxin and Shiga-like toxins on eukaryotic cells. Microbes Infect 2001, 3, 493–507. [Google Scholar] [CrossRef]
  39. Fontaine, A.; Arondel, J.; Sansonetti, P.J. Role of Shiga toxin in the pathogenesis of bacillary dysentery, studied by using a Tox- mutant of Shigella dysenteriae 1. Infect Immun 1988, 56, 3099–3109. [Google Scholar] [CrossRef]
  40. Sansonetti, P.J.; Ryter, A.; Clerc, P.; Maurelli, A.T.; Mounier, J. Multiplication of Shigella flexneri within HeLa cells: lysis of the phagocytic vacuole and plasmid-mediated contact hemolysis. Infect Immun 1986, 51, 461–469. [Google Scholar] [CrossRef]
  41. High, N.; Mounier, J.; Prevost, M.C.; Sansonetti, P.J. IpaB of Shigella flexneri causes entry into epithelial cells and escape from the phagocytic vacuole. EMBO J 1992, 11, 1991–1999. [Google Scholar] [CrossRef] [PubMed]
  42. Skoudy, A.; Mounier, J.; Aruffo, A.; Ohayon, H.; Gounon, P.; Sansonetti, P.; Tran Van Nhieu, G. CD44 binds to the Shigella IpaB protein and participates in bacterial invasion of epithelial cells. Cell Microbiol 2000, 2, 19–33. [Google Scholar] [CrossRef] [PubMed]
  43. Lafont, F.; Tran Van Nhieu, G.; Hanada, K.; Sansonetti, P.; van der Goot, F.G. Initial steps of Shigella infection depend on the cholesterol/sphingolipid raft-mediated CD44-IpaB interaction. EMBO J 2002, 21, 4449–4457. [Google Scholar] [CrossRef]
  44. Senerovic, L.; Tsunoda, S.P.; Goosmann, C.; Brinkmann, V.; Zychlinsky, A.; Meissner, F.; Kolbe, M. Spontaneous formation of IpaB ion channels in host cell membranes reveals how Shigella induces pyroptosis in macrophages. Cell Death Dis. 2012, 3, e384. [Google Scholar] [CrossRef]
  45. Gedde, M.M.; Higgins, D.E.; Tilney, L.G.; Portnoy, D.A. Role of listeriolysin O in cell-to-cell spread of Listeria monocytogenes. Infect Immun 2000, 68, 999–1003. [Google Scholar] [CrossRef] [PubMed]
  46. Vazquez-Boland, J.A.; Kocks, C.; Dramsi, S.; Ohayon, H.; Geoffroy, C.; Mengaud, J.; Cossart, P. Nucleotide sequence of the lecithinase operon of Listeria monocytogenes and possible role of lecithinase in cell-to-cell spread. Infect Immun 1992, 60, 219–230. [Google Scholar] [CrossRef]
  47. Tweten, R.K.; Parker, M.W.; Johnson, A.E. The cholesterol-dependent cytolysins. Curr Top Microbiol Immunol 2001, 257, 15–33. [Google Scholar] [CrossRef]
  48. Dal Peraro, M.; van der Goot, F.G. Pore-forming toxins: ancient, but never really out of fashion. Nat. Rev. Microbiol. 2016, 14, 77–92. [Google Scholar] [CrossRef]
  49. Geoffroy, C.; Gaillard, J.L.; Alouf, J.E.; Berche, P. Purification, characterization, and toxicity of the sulfhydryl-activated hemolysin listeriolysin O from Listeria monocytogenes. Infect Immun 1987, 55, 1641–1646. [Google Scholar] [CrossRef]
  50. Portnoy, D.A.; Tweten, R.K.; Kehoe, M.; Bielecki, J. Capacity of listeriolysin O, streptolysin O, and perfringolysin O to mediate growth of Bacillus subtilis within mammalian cells. Infect Immun 1992, 60, 2710–2717. [Google Scholar] [CrossRef]
  51. Jones, S.; Portnoy, D.A. Characterization of Listeria monocytogenes pathogenesis in a strain expressing perfringolysin O in place of listeriolysin O. Infect Immun 1994, 62, 5608–5613. [Google Scholar] [CrossRef] [PubMed]
  52. Provoda, C.J.; Lee, K.D. Bacterial pore-forming hemolysins and their use in the cytosolic delivery of macromolecules. Adv Drug Deliv Rev 2000, 41, 209–221. [Google Scholar] [CrossRef] [PubMed]
  53. Lett, M.C.; Sasakawa, C.; Okada, N.; Sakai, T.; Makino, S.; Yamada, M.; Komatsu, K.; Yoshikawa, M. virG, a plasmid-coded virulence gene of Shigella flexneri: identification of the virG protein and determination of the complete coding sequence. J Bacteriol 1989, 171, 353–359. [Google Scholar] [CrossRef]
  54. Vasselon, T.; Mounier, J.; Prevost, M.C.; Hellio, R.; Sansonetti, P.J. Stress fiber-based movement of Shigella flexneri within cells. Infect Immun 1991, 59, 1723–1732. [Google Scholar] [CrossRef]
  55. Fujita, N.; Morita, E.; Itoh, T.; Tanaka, A.; Nakaoka, M.; Osada, Y.; Umemoto, T.; Saitoh, T.; Nakatogawa, H.; Kobayashi, S.; et al. Recruitment of the autophagic machinery to endosomes during infection is mediated by ubiquitin. J Cell Biol 2013, 203, 115–128. [Google Scholar] [CrossRef] [PubMed]
  56. Maejima, I.; Takahashi, A.; Omori, H.; Kimura, T.; Takabatake, Y.; Saitoh, T.; Yamamoto, A.; Hamasaki, M.; Noda, T.; Isaka, Y.; et al. Autophagy sequesters damaged lysosomes to control lysosomal biogenesis and kidney injury. EMBO J 2013, 32, 2336–2347. [Google Scholar] [CrossRef]
  57. Dupont, N.; Lacas-Gervais, S.; Bertout, J.; Paz, I.; Freche, B.; Van Nhieu, G.T.; van der Goot, F.G.; Sansonetti, P.J.; Lafont, F. Shigella phagocytic vacuolar membrane remnants participate in the cellular response to pathogen invasion and are regulated by autophagy. Cell Host Microbe 2009, 6, 137–149. [Google Scholar] [CrossRef]
  58. Kofoed, E.M.; Vance, R.E. Innate immune recognition of bacterial ligands by NAIPs determines inflammasome specificity. Nature 2011, 477, 592–595. [Google Scholar] [CrossRef]
  59. Zhao, Y.; Yang, J.; Shi, J.; Gong, Y.N.; Lu, Q.; Xu, H.; Liu, L.; Shao, F. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus. Nature 2011, 477, 596–600. [Google Scholar] [CrossRef]
  60. Paz, I.; Sachse, M.; Dupont, N.; Mounier, J.; Cederfur, C.; Enninga, J.; Leffler, H.; Poirier, F.; Prevost, M.C.; Lafont, F.; et al. Galectin-3, a marker for vacuole lysis by invasive pathogens. Cell Microbiol 2010, 12, 530–544. [Google Scholar] [CrossRef]
  61. Marino, K.V.; Cagnoni, A.J.; Croci, D.O.; Rabinovich, G.A. Targeting galectin-driven regulatory circuits in cancer and fibrosis. Nat. Rev. Drug Discov. 2023, 22, 295–316. [Google Scholar] [CrossRef] [PubMed]
  62. Troncoso, M.F.; Elola, M.T.; Blidner, A.G.; Sarrias, L.; Espelt, M.V.; Rabinovich, G.A. The universe of galectin-binding partners and their functions in health and disease. J Biol Chem 2023, 299, 105400. [Google Scholar] [CrossRef] [PubMed]
  63. Yonekawa, Y.; Oikawa, K.; Bayarkhuu, B.; Kobayashi, K.; Saito, N.; Oikawa, I.; Yamada, R.; Chen, Y.H.; Oyanagi, K.; Shibasaki, Y.; et al. Magnetic control of membrane damage in early endosomes using internalized magnetic nanoparticles. Cell Struct Funct 2025, 50, 25–39. [Google Scholar] [CrossRef] [PubMed]
  64. Repnik, U.; Borg Distefano, M.; Speth, M.T.; Ng, M.Y.W.; Progida, C.; Hoflack, B.; Gruenberg, J.; Griffiths, G. L-leucyl-L-leucine methyl ester does not release cysteine cathepsins to the cytosol but inactivates them in transiently permeabilized lysosomes. J Cell Sci 2017, 130, 3124–3140. [Google Scholar] [CrossRef]
  65. Bjorkoy, G.; Lamark, T.; Johansen, T. p62/SQSTM1: a missing link between protein aggregates and the autophagy machinery. Autophagy 2006, 2, 138–139. [Google Scholar] [CrossRef]
  66. Seibenhener, M.L.; Babu, J.R.; Geetha, T.; Wong, H.C.; Krishna, N.R.; Wooten, M.W. Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in ubiquitin proteasome degradation. Mol Cell Biol 2004, 24, 8055–8068. [Google Scholar] [CrossRef]
  67. Koerver, L.; Papadopoulos, C.; Liu, B.; Kravic, B.; Rota, G.; Brecht, L.; Veenendaal, T.; Polajnar, M.; Bluemke, A.; Ehrmann, M.; et al. The ubiquitin-conjugating enzyme UBE2QL1 coordinates lysophagy in response to endolysosomal damage. EMBO Rep 2019, 20, e48014. [Google Scholar] [CrossRef]
  68. Papadopoulos, C.; Kirchner, P.; Bug, M.; Grum, D.; Koerver, L.; Schulze, N.; Poehler, R.; Dressler, A.; Fengler, S.; Arhzaouy, K.; et al. VCP/p97 cooperates with YOD1, UBXD1 and PLAA to drive clearance of ruptured lysosomes by autophagy. EMBO J 2017, 36, 135–150. [Google Scholar] [CrossRef]
  69. Chauhan, S.; Kumar, S.; Jain, A.; Ponpuak, M.; Mudd, M.H.; Kimura, T.; Choi, S.W.; Peters, R.; Mandell, M.; Bruun, J.A.; et al. TRIMs and Galectins Globally Cooperate and TRIM16 and Galectin-3 Co-direct Autophagy in Endomembrane Damage Homeostasis. Dev. Cell 2016, 39, 13–27. [Google Scholar] [CrossRef]
  70. Fujita, N.; Itoh, T.; Omori, H.; Fukuda, M.; Noda, T.; Yoshimori, T. The Atg16L complex specifies the site of LC3 lipidation for membrane biogenesis in autophagy. Mol Biol Cell 2008, 19, 2092–2100. [Google Scholar] [CrossRef]
  71. Jia, J.; Claude-Taupin, A.; Gu, Y.; Choi, S.W.; Peters, R.; Bissa, B.; Mudd, M.H.; Allers, L.; Pallikkuth, S.; Lidke, K.A.; et al. Galectin-3 Coordinates a Cellular System for Lysosomal Repair and Removal. Dev. Cell 2020, 52, 69–87 e68. [Google Scholar] [CrossRef]
  72. Weng, I.C.; Chen, H.L.; Lo, T.H.; Lin, W.H.; Chen, H.Y.; Hsu, D.K.; Liu, F.T. Cytosolic galectin-3 and -8 regulate antibacterial autophagy through differential recognition of host glycans on damaged phagosomes. Glycobiology 2018, 28, 392–405. [Google Scholar] [CrossRef]
  73. Thurston, T.L.; Wandel, M.P.; von Muhlinen, N.; Foeglein, A.; Randow, F. Galectin 8 targets damaged vesicles for autophagy to defend cells against bacterial invasion. Nature 2012, 482, 414–418. [Google Scholar] [CrossRef]
  74. Morishita, H.; Mizushima, N. Diverse Cellular Roles of Autophagy. Annu Rev Cell Dev Biol 2019, 35, 453–475. [Google Scholar] [CrossRef] [PubMed]
  75. Du Rietz, H.; Hedlund, H.; Wilhelmson, S.; Nordenfelt, P.; Wittrup, A. Imaging small molecule-induced endosomal escape of siRNA. Nat Commun 2020, 11, 1809. [Google Scholar] [CrossRef]
  76. Munson, M.J.; O'Driscoll, G.; Silva, A.M.; Lazaro-Ibanez, E.; Gallud, A.; Wilson, J.T.; Collen, A.; Esbjorner, E.K.; Sabirsh, A. A high-throughput Galectin-9 imaging assay for quantifying nanoparticle uptake, endosomal escape and functional RNA delivery. Commun Biol 2021, 4, 211. [Google Scholar] [CrossRef]
  77. Jia, J.; Bissa, B.; Brecht, L.; Allers, L.; Choi, S.W.; Gu, Y.; Zbinden, M.; Burge, M.R.; Timmins, G.; Hallows, K.; et al. AMPK, a Regulator of Metabolism and Autophagy, Is Activated by Lysosomal Damage via a Novel Galectin-Directed Ubiquitin Signal Transduction System. Mol Cell 2020, 77, 951–969 e959. [Google Scholar] [CrossRef]
  78. Zhu, C.; Anderson, A.C.; Schubart, A.; Xiong, H.; Imitola, J.; Khoury, S.J.; Zheng, X.X.; Strom, T.B.; Kuchroo, V.K. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat. Immunol. 2005, 6, 1245–1252. [Google Scholar] [CrossRef]
  79. Elahi, S.; Niki, T.; Hirashima, M.; Horton, H. Galectin-9 binding to Tim-3 renders activated human CD4+ T cells less susceptible to HIV-1 infection. Blood 2012, 119, 4192–4204. [Google Scholar] [CrossRef] [PubMed]
  80. Katzmann, D.J.; Stefan, C.J.; Babst, M.; Emr, S.D. Vps27 recruits ESCRT machinery to endosomes during MVB sorting. J Cell Biol 2003, 162, 413–423. [Google Scholar] [CrossRef] [PubMed]
  81. Katzmann, D.J.; Babst, M.; Emr, S.D. Ubiquitin-dependent sorting into the multivesicular body pathway requires the function of a conserved endosomal protein sorting complex, ESCRT-I. Cell 2001, 106, 145–155. [Google Scholar] [CrossRef]
  82. Babst, M.; Katzmann, D.J.; Snyder, W.B.; Wendland, B.; Emr, S.D. Endosome-associated complex, ESCRT-II, recruits transport machinery for protein sorting at the multivesicular body. Dev. Cell 2002, 3, 283–289. [Google Scholar] [CrossRef]
  83. Babst, M.; Katzmann, D.J.; Estepa-Sabal, E.J.; Meerloo, T.; Emr, S.D. Escrt-III: an endosome-associated heterooligomeric protein complex required for mvb sorting. Dev. Cell 2002, 3, 271–282. [Google Scholar] [CrossRef]
  84. Odorizzi, G.; Katzmann, D.J.; Babst, M.; Audhya, A.; Emr, S.D. Bro1 is an endosome-associated protein that functions in the MVB pathway in Saccharomyces cerevisiae. J Cell Sci 2003, 116, 1893–1903. [Google Scholar] [CrossRef]
  85. Williams, R.L.; Urbe, S. The emerging shape of the ESCRT machinery. Nat. Rev. Mol. Cell Biol. 2007, 8, 355–368. [Google Scholar] [CrossRef]
  86. Citri, A.; Yarden, Y. EGF-ERBB signalling: towards the systems level. Nat. Rev. Mol. Cell Biol. 2006, 7, 505–516. [Google Scholar] [CrossRef]
  87. von Zastrow, M.; Sorkin, A. Mechanisms for Regulating and Organizing Receptor Signaling by Endocytosis. Annu Rev Biochem 2021, 90, 709–737. [Google Scholar] [CrossRef]
  88. Tomas, A.; Futter, C.E.; Eden, E.R. EGF receptor trafficking: consequences for signaling and cancer. Trends Cell Biol. 2014, 24, 26–34. [Google Scholar] [CrossRef] [PubMed]
  89. Haglund, K.; Shimokawa, N.; Szymkiewicz, I.; Dikic, I. Cbl-directed monoubiquitination of CIN85 is involved in regulation of ligand-induced degradation of EGF receptors. Proc Natl Acad Sci U S A 2002, 99, 12191–12196. [Google Scholar] [CrossRef] [PubMed]
  90. Petrelli, A.; Gilestro, G.F.; Lanzardo, S.; Comoglio, P.M.; Migone, N.; Giordano, S. The endophilin-CIN85-Cbl complex mediates ligand-dependent downregulation of c-Met. Nature 2002, 416, 187–190. [Google Scholar] [CrossRef] [PubMed]
  91. Szymkiewicz, I.; Kowanetz, K.; Soubeyran, P.; Dinarina, A.; Lipkowitz, S.; Dikic, I. CIN85 participates in Cbl-b-mediated down-regulation of receptor tyrosine kinases. J Biol Chem 2002, 277, 39666–39672. [Google Scholar] [CrossRef] [PubMed]
  92. Soubeyran, P.; Kowanetz, K.; Szymkiewicz, I.; Langdon, W.Y.; Dikic, I. Cbl-CIN85-endophilin complex mediates ligand-induced downregulation of EGF receptors. Nature 2002, 416, 183–187. [Google Scholar] [CrossRef] [PubMed]
  93. Huang, F.; Zeng, X.; Kim, W.; Balasubramani, M.; Fortian, A.; Gygi, S.P.; Yates, N.A.; Sorkin, A. Lysine 63-linked polyubiquitination is required for EGF receptor degradation. Proc Natl Acad Sci U S A 2013, 110, 15722–15727. [Google Scholar] [CrossRef]
  94. Katzmann, D.J.; Odorizzi, G.; Emr, S.D. Receptor downregulation and multivesicular-body sorting. Nat. Rev. Mol. Cell Biol. 2002, 3, 893–905. [Google Scholar] [CrossRef]
  95. Vietri, M.; Radulovic, M.; Stenmark, H. The many functions of ESCRTs. Nat. Rev. Mol. Cell Biol. 2020, 21, 25–42. [Google Scholar] [CrossRef]
  96. Sorkin, A.; McClure, M.; Huang, F.; Carter, R. Interaction of EGF receptor and grb2 in living cells visualized by fluorescence resonance energy transfer (FRET) microscopy. Curr Biol 2000, 10, 1395–1398. [Google Scholar] [CrossRef]
  97. Surve, S.; Watkins, S.C.; Sorkin, A. EGFR-RAS-MAPK signaling is confined to the plasma membrane and associated endorecycling protrusions. J Cell Biol 2021, 220. [Google Scholar] [CrossRef]
  98. Hanson, P.I.; Roth, R.; Lin, Y.; Heuser, J.E. Plasma membrane deformation by circular arrays of ESCRT-III protein filaments. J Cell Biol 2008, 180, 389–402. [Google Scholar] [CrossRef]
  99. Wollert, T.; Wunder, C.; Lippincott-Schwartz, J.; Hurley, J.H. Membrane scission by the ESCRT-III complex. Nature 2009, 458, 172–177. [Google Scholar] [CrossRef]
  100. Elia, N.; Sougrat, R.; Spurlin, T.A.; Hurley, J.H.; Lippincott-Schwartz, J. Dynamics of endosomal sorting complex required for transport (ESCRT) machinery during cytokinesis and its role in abscission. Proc Natl Acad Sci U S A 2011, 108, 4846–4851. [Google Scholar] [CrossRef] [PubMed]
  101. Shen, Q.T.; Schuh, A.L.; Zheng, Y.; Quinney, K.; Wang, L.; Hanna, M.; Mitchell, J.C.; Otegui, M.S.; Ahlquist, P.; Cui, Q.; et al. Structural analysis and modeling reveals new mechanisms governing ESCRT-III spiral filament assembly. J Cell Biol 2014, 206, 763–777. [Google Scholar] [CrossRef]
  102. Adell, M.A.Y.; Migliano, S.M.; Upadhyayula, S.; Bykov, Y.S.; Sprenger, S.; Pakdel, M.; Vogel, G.F.; Jih, G.; Skillern, W.; Behrouzi, R.; et al. Recruitment dynamics of ESCRT-III and Vps4 to endosomes and implications for reverse membrane budding. Elife 2017, 6. [Google Scholar] [CrossRef]
  103. Jimenez, A.J.; Maiuri, P.; Lafaurie-Janvore, J.; Divoux, S.; Piel, M.; Perez, F. ESCRT machinery is required for plasma membrane repair. Science 2014, 343, 1247136. [Google Scholar] [CrossRef]
  104. Katoh, K.; Shibata, H.; Suzuki, H.; Nara, A.; Ishidoh, K.; Kominami, E.; Yoshimori, T.; Maki, M. The ALG-2-interacting protein Alix associates with CHMP4b, a human homologue of yeast Snf7 that is involved in multivesicular body sorting. J Biol Chem 2003, 278, 39104–39113. [Google Scholar] [CrossRef]
  105. von Schwedler, U.K.; Stuchell, M.; Muller, B.; Ward, D.M.; Chung, H.Y.; Morita, E.; Wang, H.E.; Davis, T.; He, G.P.; Cimbora, D.M.; et al. The protein network of HIV budding. Cell 2003, 114, 701–713. [Google Scholar] [CrossRef]
  106. Strack, B.; Calistri, A.; Craig, S.; Popova, E.; Gottlinger, H.G. AIP1/ALIX is a binding partner for HIV-1 p6 and EIAV p9 functioning in virus budding. Cell 2003, 114, 689–699. [Google Scholar] [CrossRef]
  107. Martin-Serrano, J.; Yarovoy, A.; Perez-Caballero, D.; Bieniasz, P.D. Divergent retroviral late-budding domains recruit vacuolar protein sorting factors by using alternative adaptor proteins. Proc Natl Acad Sci U S A 2003, 100, 12414–12419. [Google Scholar] [CrossRef]
  108. Scheffer, L.L.; Sreetama, S.C.; Sharma, N.; Medikayala, S.; Brown, K.J.; Defour, A.; Jaiswal, J.K. Mechanism of Ca(2)(+)-triggered ESCRT assembly and regulation of cell membrane repair. Nat Commun 2014, 5, 5646. [Google Scholar] [CrossRef] [PubMed]
  109. Vito, P.; Pellegrini, L.; Guiet, C.; D'Adamio, L. Cloning of AIP1, a novel protein that associates with the apoptosis-linked gene ALG-2 in a Ca2+-dependent reaction. J Biol Chem 1999, 274, 1533–1540. [Google Scholar] [CrossRef] [PubMed]
  110. Missotten, M.; Nichols, A.; Rieger, K.; Sadoul, R. Alix, a novel mouse protein undergoing calcium-dependent interaction with the apoptosis-linked-gene 2 (ALG-2) protein. Cell Death Differ 1999, 6, 124–129. [Google Scholar] [CrossRef] [PubMed]
  111. Trioulier, Y.; Torch, S.; Blot, B.; Cristina, N.; Chatellard-Causse, C.; Verna, J.M.; Sadoul, R. Alix, a protein regulating endosomal trafficking, is involved in neuronal death. J Biol Chem 2004, 279, 2046–2052. [Google Scholar] [CrossRef]
  112. la Cour, J.M.; Mollerup, J.; Berchtold, M.W. ALG-2 oscillates in subcellular localization, unitemporally with calcium oscillations. Biochem Biophys Res Commun 2007, 353, 1063–1067. [Google Scholar] [CrossRef]
  113. Suzuki, H.; Kawasaki, M.; Inuzuka, T.; Okumura, M.; Kakiuchi, T.; Shibata, H.; Wakatsuki, S.; Maki, M. Structural basis for Ca2+ -dependent formation of ALG-2/Alix peptide complex: Ca2+/EF3-driven arginine switch mechanism. Structure 2008, 16, 1562–1573. [Google Scholar] [CrossRef]
  114. Matsuo, H.; Chevallier, J.; Mayran, N.; Le Blanc, I.; Ferguson, C.; Faure, J.; Blanc, N.S.; Matile, S.; Dubochet, J.; Sadoul, R.; et al. Role of LBPA and Alix in multivesicular liposome formation and endosome organization. Science 2004, 303, 531–534. [Google Scholar] [CrossRef]
  115. Bissig, C.; Lenoir, M.; Velluz, M.C.; Kufareva, I.; Abagyan, R.; Overduin, M.; Gruenberg, J. Viral infection controlled by a calcium-dependent lipid-binding module in ALIX. Dev. Cell 2013, 25, 364–373. [Google Scholar] [CrossRef] [PubMed]
  116. Larios, J.; Mercier, V.; Roux, A.; Gruenberg, J. ALIX- and ESCRT-III-dependent sorting of tetraspanins to exosomes. J Cell Biol 2020, 219. [Google Scholar] [CrossRef] [PubMed]
  117. Claude-Taupin, A.; Jia, J.; Bhujabal, Z.; Garfa-Traore, M.; Kumar, S.; da Silva, G.P.D.; Javed, R.; Gu, Y.; Allers, L.; Peters, R.; et al. ATG9A protects the plasma membrane from programmed and incidental permeabilization. Nat Cell Biol 2021, 23, 846–858. [Google Scholar] [CrossRef] [PubMed]
  118. Okumura, M.; Takahashi, T.; Shibata, H.; Maki, M. Mammalian ESCRT-III-related protein IST1 has a distinctive met-pro repeat sequence that is essential for interaction with ALG-2 in the presence of Ca2+. Biosci Biotechnol Biochem 2013, 77, 1049–1054. [Google Scholar] [CrossRef] [PubMed]
  119. Radulovic, M.; Schink, K.O.; Wenzel, E.M.; Nahse, V.; Bongiovanni, A.; Lafont, F.; Stenmark, H. ESCRT-mediated lysosome repair precedes lysophagy and promotes cell survival. EMBO J 2018, 37. [Google Scholar] [CrossRef]
  120. Skowyra, M.L.; Schlesinger, P.H.; Naismith, T.V.; Hanson, P.I. Triggered recruitment of ESCRT machinery promotes endolysosomal repair. Science 2018, 360. [Google Scholar] [CrossRef]
  121. Jadot, M.; Colmant, C.; Wattiaux-De Coninck, S.; Wattiaux, R. Intralysosomal hydrolysis of glycyl-L-phenylalanine 2-naphthylamide. Biochem J 1984, 219, 965–970. [Google Scholar] [CrossRef]
  122. Berg, T.O.; Stromhaug, E.; Lovdal, T.; Seglen, O.; Berg, T. Use of glycyl-L-phenylalanine 2-naphthylamide, a lysosome-disrupting cathepsin C substrate, to distinguish between lysosomes and prelysosomal endocytic vacuoles. Biochem J 1994, 300 ( Pt 1), 229–236. [Google Scholar] [CrossRef] [PubMed]
  123. Berg, T.O.; Stromhaug, P.E.; Berg, T.; Seglen, P.O. Separation of lysosomes and autophagosomes by means of glycyl-phenylalanine-naphthylamide, a lysosome-disrupting cathepsin-C substrate. Eur J Biochem 1994, 221, 595–602. [Google Scholar] [CrossRef] [PubMed]
  124. Chen, H.Y.; Fermin, A.; Vardhana, S.; Weng, I.C.; Lo, K.F.; Chang, E.Y.; Maverakis, E.; Yang, R.Y.; Hsu, D.K.; Dustin, M.L.; et al. Galectin-3 negatively regulates TCR-mediated CD4+ T-cell activation at the immunological synapse. Proc Natl Acad Sci U S A 2009, 106, 14496–14501. [Google Scholar] [CrossRef]
  125. Wang, S.F.; Tsao, C.H.; Lin, Y.T.; Hsu, D.K.; Chiang, M.L.; Lo, C.H.; Chien, F.C.; Chen, P.; Arthur Chen, Y.M.; Chen, H.Y.; et al. Galectin-3 promotes HIV-1 budding via association with Alix and Gag p6. Glycobiology 2014, 24, 1022–1035. [Google Scholar] [CrossRef]
  126. Gahlot, P.; Kravic, B.; Rota, G.; van den Boom, J.; Levantovsky, S.; Schulze, N.; Maspero, E.; Polo, S.; Behrends, C.; Meyer, H. Lysosomal damage sensing and lysophagy initiation by SPG20-ITCH. Mol Cell 2024, 84, 1556–1569 e1510. [Google Scholar] [CrossRef] [PubMed]
  127. Mercier, V.; Larios, J.; Molinard, G.; Goujon, A.; Matile, S.; Gruenberg, J.; Roux, A. Endosomal membrane tension regulates ESCRT-III-dependent intra-lumenal vesicle formation. Nat Cell Biol 2020, 22, 947–959. [Google Scholar] [CrossRef]
  128. Dal Molin, M.; Verolet, Q.; Colom, A.; Letrun, R.; Derivery, E.; Gonzalez-Gaitan, M.; Vauthey, E.; Roux, A.; Sakai, N.; Matile, S. Fluorescent flippers for mechanosensitive membrane probes. J Am Chem Soc 2015, 137, 568–571. [Google Scholar] [CrossRef]
  129. Goujon, A.; Colom, A.; Strakova, K.; Mercier, V.; Mahecic, D.; Manley, S.; Sakai, N.; Roux, A.; Matile, S. Mechanosensitive Fluorescent Probes to Image Membrane Tension in Mitochondria, Endoplasmic Reticulum, and Lysosomes. J Am Chem Soc 2019, 141, 3380–3384. [Google Scholar] [CrossRef]
  130. Goser, V.; Kehl, A.; Roder, J.; Hensel, M. Role of the ESCRT-III complex in controlling integrity of the Salmonella-containing vacuole. Cell Microbiol 2020, 22, e13176. [Google Scholar] [CrossRef]
  131. Simons, K.; Gerl, M.J. Revitalizing membrane rafts: new tools and insights. Nat. Rev. Mol. Cell Biol. 2010, 11, 688–699. [Google Scholar] [CrossRef] [PubMed]
  132. Sanchez, S.A.; Gunther, G.; Tricerri, M.A.; Gratton, E. Methyl-beta-cyclodextrins preferentially remove cholesterol from the liquid disordered phase in giant unilamellar vesicles. J Membr Biol 2011, 241, 1–10. [Google Scholar] [CrossRef] [PubMed]
  133. Futerman, A.H.; Stieger, B.; Hubbard, A.L.; Pagano, R.E. Sphingomyelin synthesis in rat liver occurs predominantly at the cis and medial cisternae of the Golgi apparatus. J Biol Chem 1990, 265, 8650–8657. [Google Scholar] [CrossRef]
  134. Koval, M.; Pagano, R.E. Intracellular transport and metabolism of sphingomyelin. Biochim Biophys Acta 1991, 1082, 113–125. [Google Scholar] [CrossRef]
  135. Hannun, Y.A.; Luberto, C. Ceramide in the eukaryotic stress response. Trends Cell Biol. 2000, 10, 73–80. [Google Scholar] [CrossRef]
  136. Taniguchi, M.; Okazaki, T. The role of sphingomyelin and sphingomyelin synthases in cell death, proliferation and migration-from cell and animal models to human disorders. Biochim Biophys Acta 2014, 1841, 692–703. [Google Scholar] [CrossRef] [PubMed]
  137. Bandet, C.L.; Tan-Chen, S.; Bourron, O.; Le Stunff, H.; Hajduch, E. Sphingolipid Metabolism: New Insight into Ceramide-Induced Lipotoxicity in Muscle Cells. Int. J. Mol. Sci. 2019, 20. [Google Scholar] [CrossRef] [PubMed]
  138. Ellison, C.J.; Kukulski, W.; Boyle, K.B.; Munro, S.; Randow, F. Transbilayer Movement of Sphingomyelin Precedes Catastrophic Breakage of Enterobacteria-Containing Vacuoles. Curr Biol 2020, 30, 2974–2983 e2976. [Google Scholar] [CrossRef]
  139. Yamaji, A.; Sekizawa, Y.; Emoto, K.; Sakuraba, H.; Inoue, K.; Kobayashi, H.; Umeda, M. Lysenin, a novel sphingomyelin-specific binding protein. J Biol Chem 1998, 273, 5300–5306. [Google Scholar] [CrossRef]
  140. Bakrac, B.; Gutierrez-Aguirre, I.; Podlesek, Z.; Sonnen, A.F.; Gilbert, R.J.; Macek, P.; Lakey, J.H.; Anderluh, G. Molecular determinants of sphingomyelin specificity of a eukaryotic pore-forming toxin. J Biol Chem 2008, 283, 18665–18677. [Google Scholar] [CrossRef]
  141. Bakrac, B.; Kladnik, A.; Macek, P.; McHaffie, G.; Werner, A.; Lakey, J.H.; Anderluh, G. A toxin-based probe reveals cytoplasmic exposure of Golgi sphingomyelin. J Biol Chem 2010, 285, 22186–22195. [Google Scholar] [CrossRef] [PubMed]
  142. Yachi, R.; Uchida, Y.; Balakrishna, B.H.; Anderluh, G.; Kobayashi, T.; Taguchi, T.; Arai, H. Subcellular localization of sphingomyelin revealed by two toxin-based probes in mammalian cells. Genes Cells 2012, 17, 720–727. [Google Scholar] [CrossRef] [PubMed]
  143. Ishitsuka, R.; Yamaji-Hasegawa, A.; Makino, A.; Hirabayashi, Y.; Kobayashi, T. A lipid-specific toxin reveals heterogeneity of sphingomyelin-containing membranes. Biophys J 2004, 86, 296–307. [Google Scholar] [CrossRef] [PubMed]
  144. Boyle, K.B.; Ellison, C.J.; Elliott, P.R.; Schuschnig, M.; Grimes, K.; Dionne, M.S.; Sasakawa, C.; Munro, S.; Martens, S.; Randow, F. TECPR1 conjugates LC3 to damaged endomembranes upon detection of sphingomyelin exposure. EMBO J 2023, 42, e113012. [Google Scholar] [CrossRef]
  145. Ogawa, M.; Yoshikawa, Y.; Kobayashi, T.; Mimuro, H.; Fukumatsu, M.; Kiga, K.; Piao, Z.; Ashida, H.; Yoshida, M.; Kakuta, S.; et al. A Tecpr1-dependent selective autophagy pathway targets bacterial pathogens. Cell Host Microbe 2011, 9, 376–389. [Google Scholar] [CrossRef]
  146. Allaoui, A.; Mounier, J.; Prevost, M.C.; Sansonetti, P.J.; Parsot, C. icsB: a Shigella flexneri virulence gene necessary for the lysis of protrusions during intercellular spread. Mol Microbiol 1992, 6, 1605–1616. [Google Scholar] [CrossRef]
  147. Niekamp, P.; Scharte, F.; Sokoya, T.; Vittadello, L.; Kim, Y.; Deng, Y.; Sudhoff, E.; Hilderink, A.; Imlau, M.; Clarke, C.J.; et al. Ca(2+)-activated sphingomyelin scrambling and turnover mediate ESCRT-independent lysosomal repair. Nat Commun 2022, 13, 1875. [Google Scholar] [CrossRef]
  148. Jia, J.; Wang, F.; Bhujabal, Z.; Peters, R.; Mudd, M.; Duque, T.; Allers, L.; Javed, R.; Salemi, M.; Behrends, C.; et al. Stress granules and mTOR are regulated by membrane atg8ylation during lysosomal damage. J Cell Biol 2022, 221. [Google Scholar] [CrossRef]
  149. Yang, P.; Mathieu, C.; Kolaitis, R.M.; Zhang, P.; Messing, J.; Yurtsever, U.; Yang, Z.; Wu, J.; Li, Y.; Pan, Q.; et al. G3BP1 Is a Tunable Switch that Triggers Phase Separation to Assemble Stress Granules. Cell 2020, 181, 325–345 e328. [Google Scholar] [CrossRef]
  150. Guillen-Boixet, J.; Kopach, A.; Holehouse, A.S.; Wittmann, S.; Jahnel, M.; Schlussler, R.; Kim, K.; Trussina, I.; Wang, J.; Mateju, D.; et al. RNA-Induced Conformational Switching and Clustering of G3BP Drive Stress Granule Assembly by Condensation. Cell 2020, 181, 346–361 e317. [Google Scholar] [CrossRef]
  151. Bussi, C.; Mangiarotti, A.; Vanhille-Campos, C.; Aylan, B.; Pellegrino, E.; Athanasiadi, N.; Fearns, A.; Rodgers, A.; Franzmann, T.M.; Saric, A.; et al. Stress granules plug and stabilize damaged endolysosomal membranes. Nature 2023, 623, 1062–1069. [Google Scholar] [CrossRef]
  152. Kedersha, N.; Chen, S.; Gilks, N.; Li, W.; Miller, I.J.; Stahl, J.; Anderson, P. Evidence that ternary complex (eIF2-GTP-tRNA(i)(Met))-deficient preinitiation complexes are core constituents of mammalian stress granules. Mol Biol Cell 2002, 13, 195–210. [Google Scholar] [CrossRef]
  153. Duran, J.; Salinas, J.E.; Wheaton, R.P.; Poolsup, S.; Allers, L.; Rosas-Lemus, M.; Chen, L.; Cheng, Q.; Pu, J.; Salemi, M.; et al. Calcium signaling from damaged lysosomes induces cytoprotective stress granules. EMBO J 2024, 43, 6410–6443. [Google Scholar] [CrossRef] [PubMed]
  154. Stenmark, H.; Vitale, G.; Ullrich, O.; Zerial, M. Rabaptin-5 is a direct effector of the small GTPase Rab5 in endocytic membrane fusion. Cell 1995, 83, 423–432. [Google Scholar] [CrossRef] [PubMed]
  155. Vitale, G.; Rybin, V.; Christoforidis, S.; Thornqvist, P.; McCaffrey, M.; Stenmark, H.; Zerial, M. Distinct Rab-binding domains mediate the interaction of Rabaptin-5 with GTP-bound Rab4 and Rab5. EMBO J 1998, 17, 1941–1951. [Google Scholar] [CrossRef]
  156. Lippe, R.; Miaczynska, M.; Rybin, V.; Runge, A.; Zerial, M. Functional synergy between Rab5 effector Rabaptin-5 and exchange factor Rabex-5 when physically associated in a complex. Mol Biol Cell 2001, 12, 2219–2228. [Google Scholar] [CrossRef] [PubMed]
  157. Shiba, Y.; Takatsu, H.; Shin, H.W.; Nakayama, K. Gamma-adaptin interacts directly with Rabaptin-5 through its ear domain. J. Biochem. 2002, 131, 327–336. [Google Scholar] [CrossRef]
  158. Millarte, V.; Schlienger, S.; Kalin, S.; Spiess, M. Rabaptin5 targets autophagy to damaged endosomes and Salmonella vacuoles via FIP200 and ATG16L1. EMBO Rep 2022, 23, e53429. [Google Scholar] [CrossRef]
  159. Nakatsu, F.; Kawasaki, A. Functions of Oxysterol-Binding Proteins at Membrane Contact Sites and Their Control by Phosphoinositide Metabolism. Front Cell Dev Biol 2021, 9, 664788. [Google Scholar] [CrossRef]
  160. Prinz, W.A.; Toulmay, A.; Balla, T. The functional universe of membrane contact sites. Nat. Rev. Mol. Cell Biol. 2020, 21, 7–24. [Google Scholar] [CrossRef]
  161. Arora, A.; Taskinen, J.H.; Olkkonen, V.M. Coordination of inter-organelle communication and lipid fluxes by OSBP-related proteins. Prog Lipid Res 2022, 86, 101146. [Google Scholar] [CrossRef]
  162. Balla, A.; Tuymetova, G.; Tsiomenko, A.; Varnai, P.; Balla, T. A plasma membrane pool of phosphatidylinositol 4-phosphate is generated by phosphatidylinositol 4-kinase type-III alpha: studies with the PH domains of the oxysterol binding protein and FAPP1. Mol Biol Cell 2005, 16, 1282–1295. [Google Scholar] [CrossRef]
  163. Hammond, G.R.; Machner, M.P.; Balla, T. A novel probe for phosphatidylinositol 4-phosphate reveals multiple pools beyond the Golgi. J Cell Biol 2014, 205, 113–126. [Google Scholar] [CrossRef] [PubMed]
  164. Dejgaard, S.Y.; Presley, J.F. Arfs on the Golgi: four conductors, one orchestra. Front Mol Biosci 2025, 12, 1612531. [Google Scholar] [CrossRef]
  165. Lippincott-Schwartz, J.; Yuan, L.C.; Bonifacino, J.S.; Klausner, R.D. Rapid redistribution of Golgi proteins into the ER in cells treated with brefeldin A: evidence for membrane cycling from Golgi to ER. Cell 1989, 56, 801–813. [Google Scholar] [CrossRef]
  166. Donaldson, J.G.; Lippincott-Schwartz, J.; Klausner, R.D. Guanine nucleotides modulate the effects of brefeldin A in semipermeable cells: regulation of the association of a 110-kD peripheral membrane protein with the Golgi apparatus. J Cell Biol 1991, 112, 579–588. [Google Scholar] [CrossRef]
  167. Niu, T.K.; Pfeifer, A.C.; Lippincott-Schwartz, J.; Jackson, C.L. Dynamics of GBF1, a Brefeldin A-sensitive Arf1 exchange factor at the Golgi. Mol Biol Cell 2005, 16, 1213–1222. [Google Scholar] [CrossRef] [PubMed]
  168. Radulovic, M.; Wenzel, E.M.; Gilani, S.; Holland, L.K.; Lystad, A.H.; Phuyal, S.; Olkkonen, V.M.; Brech, A.; Jaattela, M.; Maeda, K.; et al. Cholesterol transfer via endoplasmic reticulum contacts mediates lysosome damage repair. EMBO J 2022, 41, e112677. [Google Scholar] [CrossRef]
  169. Tan, J.X.; Finkel, T. A phosphoinositide signalling pathway mediates rapid lysosomal repair. Nature 2022, 609, 815–821. [Google Scholar] [CrossRef] [PubMed]
  170. Tsukada, M.; Ohsumi, Y. Isolation and characterization of autophagy-defective mutants of Saccharomyces cerevisiae. FEBS Lett. 1993, 333, 169–174. [Google Scholar] [CrossRef]
  171. Harding, T.M.; Morano, K.A.; Scott, S.V.; Klionsky, D.J. Isolation and characterization of yeast mutants in the cytoplasm to vacuole protein targeting pathway. J Cell Biol 1995, 131, 591–602. [Google Scholar] [CrossRef]
  172. Maeda, S.; Otomo, C.; Otomo, T. The autophagic membrane tether ATG2A transfers lipids between membranes. Elife 2019, 8. [Google Scholar] [CrossRef]
  173. Osawa, T.; Kotani, T.; Kawaoka, T.; Hirata, E.; Suzuki, K.; Nakatogawa, H.; Ohsumi, Y.; Noda, N.N. Atg2 mediates direct lipid transfer between membranes for autophagosome formation. Nat. Struct. Mol. Biol. 2019, 26, 281–288. [Google Scholar] [CrossRef]
  174. Valverde, D.P.; Yu, S.; Boggavarapu, V.; Kumar, N.; Lees, J.A.; Walz, T.; Reinisch, K.M.; Melia, T.J. ATG2 transports lipids to promote autophagosome biogenesis. J Cell Biol 2019, 218, 1787–1798. [Google Scholar] [CrossRef]
  175. Osawa, T.; Ishii, Y.; Noda, N.N. Human ATG2B possesses a lipid transfer activity which is accelerated by negatively charged lipids and WIPI4. Genes Cells 2020, 25, 65–70. [Google Scholar] [CrossRef] [PubMed]
  176. Lang, T.; Reiche, S.; Straub, M.; Bredschneider, M.; Thumm, M. Autophagy and the cvt pathway both depend on AUT9. J Bacteriol 2000, 182, 2125–2133. [Google Scholar] [CrossRef] [PubMed]
  177. Noda, T.; Kim, J.; Huang, W.P.; Baba, M.; Tokunaga, C.; Ohsumi, Y.; Klionsky, D.J. Apg9p/Cvt7p is an integral membrane protein required for transport vesicle formation in the Cvt and autophagy pathways. J Cell Biol 2000, 148, 465–480. [Google Scholar] [CrossRef] [PubMed]
  178. De Tito, S.; Almacellas, E.; Dai Yu, D.; Millard, E.; Zhang, W.; de Heus, C.; Queval, C.; Hervas, J.H.; Pellegrino, E.; Panagi, I.; et al. ATG9A and ARFIP2 cooperate to control PI4P levels for lysosomal repair. Dev. Cell 2025. [CrossRef]
  179. Young, A.R.; Chan, E.Y.; Hu, X.W.; Kochl, R.; Crawshaw, S.G.; High, S.; Hailey, D.W.; Lippincott-Schwartz, J.; Tooze, S.A. Starvation and ULK1-dependent cycling of mammalian Atg9 between the TGN and endosomes. J Cell Sci 2006, 119, 3888–3900. [Google Scholar] [CrossRef]
  180. Orsi, A.; Razi, M.; Dooley, H.C.; Robinson, D.; Weston, A.E.; Collinson, L.M.; Tooze, S.A. Dynamic and transient interactions of Atg9 with autophagosomes, but not membrane integration, are required for autophagy. Mol Biol Cell 2012, 23, 1860–1873. [Google Scholar] [CrossRef]
  181. Kageyama, S.; Omori, H.; Saitoh, T.; Sone, T.; Guan, J.L.; Akira, S.; Imamoto, F.; Noda, T.; Yoshimori, T. The LC3 recruitment mechanism is separate from Atg9L1-dependent membrane formation in the autophagic response against Salmonella. Mol Biol Cell 2011, 22, 2290–2300. [Google Scholar] [CrossRef]
  182. Guardia, C.M.; Tan, X.F.; Lian, T.; Rana, M.S.; Zhou, W.; Christenson, E.T.; Lowry, A.J.; Faraldo-Gomez, J.D.; Bonifacino, J.S.; Jiang, J.; et al. Structure of Human ATG9A, the Only Transmembrane Protein of the Core Autophagy Machinery. Cell Rep. 2020, 31, 107837. [Google Scholar] [CrossRef]
  183. Matoba, K.; Kotani, T.; Tsutsumi, A.; Tsuji, T.; Mori, T.; Noshiro, D.; Sugita, Y.; Nomura, N.; Iwata, S.; Ohsumi, Y.; et al. Atg9 is a lipid scramblase that mediates autophagosomal membrane expansion. Nat. Struct. Mol. Biol. 2020, 27, 1185–1193. [Google Scholar] [CrossRef]
  184. van Vliet, A.R.; Chiduza, G.N.; Maslen, S.L.; Pye, V.E.; Joshi, D.; De Tito, S.; Jefferies, H.B.J.; Christodoulou, E.; Roustan, C.; Punch, E.; et al. ATG9A and ATG2A form a heteromeric complex essential for autophagosome formation. Mol Cell 2022, 82, 4324–4339 e4328. [Google Scholar] [CrossRef]
  185. Popovic, D.; Dikic, I. TBC1D5 and the AP2 complex regulate ATG9 trafficking and initiation of autophagy. EMBO Rep 2014, 15, 392–401. [Google Scholar] [CrossRef] [PubMed]
  186. Imai, K.; Hao, F.; Fujita, N.; Tsuji, Y.; Oe, Y.; Araki, Y.; Hamasaki, M.; Noda, T.; Yoshimori, T. Atg9A trafficking through the recycling endosomes is required for autophagosome formation. J Cell Sci 2016, 129, 3781–3791. [Google Scholar] [CrossRef] [PubMed]
  187. Mattera, R.; Park, S.Y.; De Pace, R.; Guardia, C.M.; Bonifacino, J.S. AP-4 mediates export of ATG9A from the trans-Golgi network to promote autophagosome formation. Proc Natl Acad Sci U S A 2017, 114, E10697–E10706. [Google Scholar] [CrossRef] [PubMed]
  188. Davies, A.K.; Itzhak, D.N.; Edgar, J.R.; Archuleta, T.L.; Hirst, J.; Jackson, L.P.; Robinson, M.S.; Borner, G.H.H. AP-4 vesicles contribute to spatial control of autophagy via RUSC-dependent peripheral delivery of ATG9A. Nat Commun 2018, 9, 3958. [Google Scholar] [CrossRef]
  189. Jia, S.; Wang, Y.; You, Z.; Liu, B.; Gao, J.; Liu, W. Mammalian Atg9 contributes to the post-Golgi transport of lysosomal hydrolases by interacting with adaptor protein-1. FEBS Lett. 2017, 591, 4027–4038. [Google Scholar] [CrossRef]
  190. Cruz-Garcia, D.; Ortega-Bellido, M.; Scarpa, M.; Villeneuve, J.; Jovic, M.; Porzner, M.; Balla, T.; Seufferlein, T.; Malhotra, V. Recruitment of arfaptins to the trans-Golgi network by PI(4)P and their involvement in cargo export. EMBO J 2013, 32, 1717–1729. [Google Scholar] [CrossRef]
  191. Moss, S.E.; Morgan, R.O. The annexins. Genome Biol 2004, 5, 219. [Google Scholar] [CrossRef]
  192. Gerke, V.; Creutz, C.E.; Moss, S.E. Annexins: linking Ca2+ signalling to membrane dynamics. Nat. Rev. Mol. Cell Biol. 2005, 6, 449–461. [Google Scholar] [CrossRef]
  193. Gerke, V. Consensus peptide antibodies reveal a widespread occurrence of Ca2+/lipid-binding proteins of the annexin family. FEBS Lett. 1989, 258, 259–262. [Google Scholar] [CrossRef]
  194. Blackwood, R.A.; Ernst, J.D. Characterization of Ca2(+)-dependent phospholipid binding, vesicle aggregation and membrane fusion by annexins. Biochem J 1990, 266, 195–200. [Google Scholar] [CrossRef]
  195. Meers, P.; Mealy, T. Calcium-dependent annexin V binding to phospholipids: stoichiometry, specificity, and the role of negative charge. Biochemistry 1993, 32, 11711–11721. [Google Scholar] [CrossRef] [PubMed]
  196. Junker, M.; Creutz, C.E. Ca(2+)-dependent binding of endonexin (annexin IV) to membranes: analysis of the effects of membrane lipid composition and development of a predictive model for the binding interaction. Biochemistry 1994, 33, 8930–8940. [Google Scholar] [CrossRef] [PubMed]
  197. Tokumitsu, H.; Mizutani, A.; Minami, H.; Kobayashi, R.; Hidaka, H. A calcyclin-associated protein is a newly identified member of the Ca2+/phospholipid-binding proteins, annexin family. J Biol Chem 1992, 267, 8919–8924. [Google Scholar] [CrossRef] [PubMed]
  198. Hayes, M.J.; Merrifield, C.J.; Shao, D.; Ayala-Sanmartin, J.; Schorey, C.D.; Levine, T.P.; Proust, J.; Curran, J.; Bailly, M.; Moss, S.E. Annexin 2 binding to phosphatidylinositol 4,5-bisphosphate on endocytic vesicles is regulated by the stress response pathway. J Biol Chem 2004, 279, 14157–14164. [Google Scholar] [CrossRef]
  199. Rescher, U.; Ruhe, D.; Ludwig, C.; Zobiack, N.; Gerke, V. Annexin 2 is a phosphatidylinositol (4,5)-bisphosphate binding protein recruited to actin assembly sites at cellular membranes. J Cell Sci 2004, 117, 3473–3480. [Google Scholar] [CrossRef]
  200. Harder, T.; Kellner, R.; Parton, R.G.; Gruenberg, J. Specific release of membrane-bound annexin II and cortical cytoskeletal elements by sequestration of membrane cholesterol. Mol Biol Cell 1997, 8, 533–545. [Google Scholar] [CrossRef]
  201. Lennon, N.J.; Kho, A.; Bacskai, B.J.; Perlmutter, S.L.; Hyman, B.T.; Brown, R.H., Jr. Dysferlin interacts with annexins A1 and A2 and mediates sarcolemmal wound-healing. J Biol Chem 2003, 278, 50466–50473. [Google Scholar] [CrossRef]
  202. Bittel, D.C.; Chandra, G.; Tirunagri, L.M.S.; Deora, A.B.; Medikayala, S.; Scheffer, L.; Defour, A.; Jaiswal, J.K. Annexin A2 Mediates Dysferlin Accumulation and Muscle Cell Membrane Repair. Cells 2020, 9. [Google Scholar] [CrossRef]
  203. McNeil, A.K.; Rescher, U.; Gerke, V.; McNeil, P.L. Requirement for annexin A1 in plasma membrane repair. J Biol Chem 2006, 281, 35202–35207. [Google Scholar] [CrossRef]
  204. Boye, T.L.; Maeda, K.; Pezeshkian, W.; Sonder, S.L.; Haeger, S.C.; Gerke, V.; Simonsen, A.C.; Nylandsted, J. Annexin A4 and A6 induce membrane curvature and constriction during cell membrane repair. Nat Commun 2017, 8, 1623. [Google Scholar] [CrossRef]
  205. Bouter, A.; Gounou, C.; Berat, R.; Tan, S.; Gallois, B.; Granier, T.; d'Estaintot, B.L.; Poschl, E.; Brachvogel, B.; Brisson, A.R. Annexin-A5 assembled into two-dimensional arrays promotes cell membrane repair. Nat Commun 2011, 2, 270. [Google Scholar] [CrossRef] [PubMed]
  206. Sonder, S.L.; Boye, T.L.; Tolle, R.; Dengjel, J.; Maeda, K.; Jaattela, M.; Simonsen, A.C.; Jaiswal, J.K.; Nylandsted, J. Annexin A7 is required for ESCRT III-mediated plasma membrane repair. Sci. Rep. 2019, 9, 6726. [Google Scholar] [CrossRef]
  207. Koerdt, S.N.; Gerke, V. Annexin A2 is involved in Ca(2+)-dependent plasma membrane repair in primary human endothelial cells. Biochim Biophys Acta Mol Cell Res 2017, 1864, 1046–1053. [Google Scholar] [CrossRef]
  208. Demonbreun, A.R.; Quattrocelli, M.; Barefield, D.Y.; Allen, M.V.; Swanson, K.E.; McNally, E.M. An actin-dependent annexin complex mediates plasma membrane repair in muscle. J Cell Biol 2016, 213, 705–718. [Google Scholar] [CrossRef] [PubMed]
  209. Scharf, B.; Clement, C.C.; Wu, X.X.; Morozova, K.; Zanolini, D.; Follenzi, A.; Larocca, J.N.; Levon, K.; Sutterwala, F.S.; Rand, J.; et al. Annexin A2 binds to endosomes following organelle destabilization by particulate wear debris. Nat Commun 2012, 3, 755. [Google Scholar] [CrossRef] [PubMed]
  210. Maitra, R.; Clement, C.C.; Scharf, B.; Crisi, G.M.; Chitta, S.; Paget, D.; Purdue, P.E.; Cobelli, N.; Santambrogio, L. Endosomal damage and TLR2 mediated inflammasome activation by alkane particles in the generation of aseptic osteolysis. Mol Immunol 2009, 47, 175–184. [Google Scholar] [CrossRef] [PubMed]
  211. Yim, W.W.; Yamamoto, H.; Mizushima, N. Annexins A1 and A2 are recruited to larger lysosomal injuries independently of ESCRTs to promote repair. FEBS Lett. 2022, 596, 991–1003. [Google Scholar] [CrossRef]
  212. Ebstrup, M.L.; Sonder, S.L.; Fogde, D.L.; Heitmann, A.S.B.; Dietrich, T.N.; Dias, C.; Jaattela, M.; Maeda, K.; Nylandsted, J. Annexin A7 mediates lysosome repair independently of ESCRT-III. Front Cell Dev Biol 2023, 11, 1211498. [Google Scholar] [CrossRef]
  213. Andrews, N.W.; Almeida, P.E.; Corrotte, M. Damage control: cellular mechanisms of plasma membrane repair. Trends Cell Biol. 2014, 24, 734–742. [Google Scholar] [CrossRef]
  214. Emans, N.; Gorvel, J.P.; Walter, C.; Gerke, V.; Kellner, R.; Griffiths, G.; Gruenberg, J. Annexin II is a major component of fusogenic endosomal vesicles. J Cell Biol 1993, 120, 1357–1369. [Google Scholar] [CrossRef] [PubMed]
  215. Chasserot-Golaz, S.; Vitale, N.; Umbrecht-Jenck, E.; Knight, D.; Gerke, V.; Bader, M.F. Annexin 2 promotes the formation of lipid microdomains required for calcium-regulated exocytosis of dense-core vesicles. Mol Biol Cell 2005, 16, 1108–1119. [Google Scholar] [CrossRef] [PubMed]
  216. Raposo, G.; Stoorvogel, W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol 2013, 200, 373–383. [Google Scholar] [CrossRef]
  217. Harding, C.; Heuser, J.; Stahl, P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol 1983, 97, 329–339. [Google Scholar] [CrossRef] [PubMed]
  218. Harding, C.; Heuser, J.; Stahl, P. Endocytosis and intracellular processing of transferrin and colloidal gold-transferrin in rat reticulocytes: demonstration of a pathway for receptor shedding. Eur J Cell Biol 1984, 35, 256–263. [Google Scholar]
  219. Pan, B.T.; Teng, K.; Wu, C.; Adam, M.; Johnstone, R.M. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol 1985, 101, 942–948. [Google Scholar] [CrossRef]
  220. Heijnen, H.F.; Schiel, A.E.; Fijnheer, R.; Geuze, H.J.; Sixma, J.J. Activated platelets release two types of membrane vesicles: microvesicles by surface shedding and exosomes derived from exocytosis of multivesicular bodies and alpha-granules. Blood 1999, 94, 3791–3799. [Google Scholar] [CrossRef]
  221. Williams, J.K.; Ngo, J.M.; Lehman, I.M.; Schekman, R. Annexin A6 mediates calcium-dependent exosome secretion during plasma membrane repair. Elife 2023, 12. [Google Scholar] [CrossRef]
  222. Ostrowski, M.; Carmo, N.B.; Krumeich, S.; Fanget, I.; Raposo, G.; Savina, A.; Moita, C.F.; Schauer, K.; Hume, A.N.; Freitas, R.P.; et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat Cell Biol 2010, 12, 19–30; sup pp 11. [Google Scholar] [CrossRef]
  223. Verweij, F.J.; Bebelman, M.P.; Jimenez, C.R.; Garcia-Vallejo, J.J.; Janssen, H.; Neefjes, J.; Knol, J.C.; de Goeij-de Haas, R.; Piersma, S.R.; Baglio, S.R.; et al. Quantifying exosome secretion from single cells reveals a modulatory role for GPCR signaling. J Cell Biol 2018, 217, 1129–1142. [Google Scholar] [CrossRef] [PubMed]
  224. Fordjour, F.K.; Guo, C.; Ai, Y.; Daaboul, G.G.; Gould, S.J. A shared, stochastic pathway mediates exosome protein budding along plasma and endosome membranes. J Biol Chem 2022, 298, 102394. [Google Scholar] [CrossRef]
  225. Edgar, J.R.; Eden, E.R.; Futter, C.E. Hrs- and CD63-dependent competing mechanisms make different sized endosomal intraluminal vesicles. Traffic 2014, 15, 197–211. [Google Scholar] [CrossRef]
  226. Suzuki, K.; Okawa, Y.; Akter, S.; Ito, H.; Shiba, Y. Arf GTPase-Activating proteins ADAP1 and ARAP1 regulate incorporation of CD63 in multivesicular bodies. Biol Open 2024, 13. [Google Scholar] [CrossRef]
  227. Shiba, Y.; Randazzo, P.A. ArfGAP1 function in COPI mediated membrane traffic: currently debated models and comparison to other coat-binding ArfGAPs. Histol Histopathol 2012, 27, 1143–1153. [Google Scholar]
  228. Shiba, Y.; Randazzo, P.A. ArfGAPs: key regulators for receptor sorting. Receptors Clin Investig 2014, 1, e158. [Google Scholar] [CrossRef] [PubMed]
  229. Tanna, C.E.; Goss, L.B.; Ludwig, C.G.; Chen, P.W. Arf GAPs as Regulators of the Actin Cytoskeleton-An Update. Int. J. Mol. Sci. 2019, 20. [Google Scholar] [CrossRef] [PubMed]
  230. Shiba, Y.; Kametaka, S.; Waguri, S.; Presley, J.F.; Randazzo, P.A. ArfGAP3 regulates the transport of cation-independent mannose 6-phosphate receptor in the post-Golgi compartment. Curr Biol 2013, 23, 1945–1951. [Google Scholar] [CrossRef]
  231. Shelke, G.V.; Williamson, C.D.; Jarnik, M.; Bonifacino, J.S. Inhibition of endolysosome fusion increases exosome secretion. J Cell Biol 2023, 222. [Google Scholar] [CrossRef] [PubMed]
  232. Verweij, F.J.; Bebelman, M.P.; George, A.E.; Couty, M.; Becot, A.; Palmulli, R.; Heiligenstein, X.; Sires-Campos, J.; Raposo, G.; Pegtel, D.M.; et al. ER membrane contact sites support endosomal small GTPase conversion for exosome secretion. J Cell Biol 2022, 221. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Shigella entry into epithelial cells.
Figure 1. Shigella entry into epithelial cells.
Preprints 178362 g001
Figure 2. Schematic model of molecules recruited to damaged organelles and artificial damaging methods.
Figure 2. Schematic model of molecules recruited to damaged organelles and artificial damaging methods.
Preprints 178362 g002
Table 1. Molecules recruited to damaged organelles.
Table 1. Molecules recruited to damaged organelles.
molecule recognition Recruitment upon membrane damage timing reference
Galectin-3 lumenal glycosylation lysosomes, endosomes 10-30min [55,56,57,60]
Galectin-8 lumenal glycosylation lysosomes, endosomes 30-60min [73]
Galectin-9 lumenal glycosylation lysosomes,endosomes 5-10min [75,76,77]
LC3   lysosomes, endosomes, autophagosomes 30-60min [55,56,57,60,73]
Ubiquitin   lysosomes, endosomes, plasma membrane 30-60min [67,103,158]
UBE2QL1   lysosomes 30-60min [67]
ALIX ALG2 binding under Ca2+, Gal-3 binding lysosomes, endosomes, plasma membrane 30sec~ [71,103,108,119,120,127]
ALG2 Ca2+ lysosomes, endosomes, plasma membrane 20sec~ [108,112,206]
CHMP4B ALIX/TSG101 lysosomes, endosomes, plasma membrane 60sec~ [71,108,119,120,127]
IST1 ALG2 binding under Ca2+ lysosomes 60sec~ [118,126]
SPG20 Peroxidized lipids, IST1 lysosomes 5min~ [126]
ITCH SPG20 binding lysosomes 15min [126]
Lysenin Sphingomyelin exposure to cytosol lysosomes, endosomes, plasma membrane 5-40min [138,139]
EqtSM Sphingomyelin exposure to cytosol lysosomes, endosomes, plasma membrane 2min~ [142,147]
TECPR1 Sphingomyelin exposure to cytosol bacteria-positive autophagosomes, lysosomes 10-30min [144,145]
G3BP1/2   Stress granules, Slightly overlap with damaged lysosomes 30sec~ [148,151,153]
Rabaptin-5 (Rab4,Rab5)a endosomes 30min [158]
PI4KIIA   lysosomes 10min [169]
SidM PI4P lysosomes 5-10min [168,178]
ORP9-11 PI4P lysosomes 10min [169]
OSBP PI4P, (Arf1)a lysosomes 6-30min [168,169]
ATG2A   lysosomes 10 min [169]
ATG9A Ca2+ lysosomes, plasma membrane 15-45min [117,178]
Annexin A1,2,4,5,6,7 Ca2+ , negative-charged lipids plasma membrane 2-50 sec [204,205,206,207,208]
Annexin A1, 2, 6,7 Ca2+ , negative-charged lipids MVBs, lysosomes, plasma membrane after fusion 10-30min [209,211,212,221]
a ( ) binding sites known in normal condition.
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.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated