Skip to main content
Advertisement
  • Loading metrics

The multifaceted interactions between pathogens and host ESCRT machinery

Abstract

The Endosomal Sorting Complex Required for Transport (ESCRT) machinery consists of multiple protein complexes that coordinate vesicle budding away from the host cytosol. ESCRTs function in many fundamental cellular processes including the biogenesis of multivesicular bodies and exosomes, membrane repair and restoration, and cell abscission during cytokinesis. Work over the past 2 decades has shown that a diverse cohort of viruses critically rely upon host ESCRT machinery for virus replication and envelopment. More recent studies reported that intracellular bacteria and the intracellular parasite Toxoplasma gondii benefit from, antagonize, or exploit host ESCRT machinery to preserve their intracellular niche, gain resources, or egress from infected cells. Here, we review how intracellular pathogens interact with the ESCRT machinery of their hosts, highlighting the variety of strategies they use to bind ESCRT complexes using short linear amino acid motifs like those used by ESCRTs to sequentially assemble on target membranes. Future work exposing new mechanisms of this molecular mimicry will yield novel insight of how pathogens exploit host ESCRT machinery and how ESCRTs facilitate key cellular processes.

Overview of the host endosomal sorting complex required for transport machinery

Endosomal Sorting Complex Required for Transport (ESCRT) machinery functions in various cellular pathways ranging from vesicular trafficking and cytokinesis to membrane repair (see reviews [16]). The ESCRT machinery (also termed ESCRTs) consists of 4 protein complexes (ESCRT-0, -I, -II, and -III), which sequentially associate with a target membrane and the AAA ATPase vacuolar protein sorting-associated proteins 4A and 4B (VPS4A/B) to mediate membrane deformation and vesicle scission away from the cytosol [7]. Their recruitment to various cellular sites is dependent on ESCRT adaptor proteins or protein complexes that initiate the machinery assembly [6]. For example, in its canonical function involving the formation of multivesicular bodies (MVBs), recruitment of ESCRTs is initiated by ESCRT-0 recognition of ubiquitinated cargo targeted for degradation [5]. Table 1 summarizes the interactions of complexes that mediate the assembly of the ESCRT machinery. However, other ESCRT functions do not require ESCRT-0 for initiation and instead depend on different adaptors for the initiation of assembly. This is the case for the role of ESCRTs in plasma membrane repair, in which the ESCRT adaptor apoptosis linked gene-2 (ALG-2, also known as PDCD6) interacts with ESCRT components for the repair of injured membranes [6]. Another example is the recruitment of the ESCRTs by the centrosomal protein of 55 kDa (CEP55) during cytokinesis [4]. Due to the multiple functions of ESCRTs in cells, many intracellular pathogens have evolved ways to exploit ESCRTs for key aspects of their life cycles including for pathogen replication, assembly, and egress. This review highlights the clever, and in some cases convergent, strategies by which pathogens make use of the host ESCRT machinery to promote their intracellular survival and propagation.

Viral late domain motifs provided first insights into how viruses exploit host ESCRTs

The first described interaction of a pathogen with the host ESCRT machinery was identified while studying retroviral budding from cells. Two decades ago, it was discovered that the human immunodeficiency virus-1 (HIV-1) requires host ESCRT components for viral budding [11]. Since then, numerous studies have shed light on the mechanisms by which this important human pathogen exploits the ESCRT machinery of its host [11,12]. A significant breakthrough was the identification of a P[T/S]AP motif in the HIV-1 Gag protein, which functions in the recruitment of the ESCRT-I component tumor susceptibility gene 101 (TSG101) [13]. By doing so, HIV-1 Gag essentially mimics the interaction between the TSG101 UEV domain and the PTAP motif in the ESCRT-0 hepatocyte growth factor-regulated tyrosine kinase substrate protein (HRS/ HGS) [1316]. This proline-rich sequence, known as a late domain motif, is encoded in the HIV-1 Gag p6 domain that functions at the late stages of the infection cycle, hence the name [17]. Although the PTAP late domain motif was first discovered in HIV-1 Gag, studies have shown that other retroviruses and nonrelated viruses like Ebola virus (EBOV) also use the PTAP late domain for the recruitment of ESCRTs for viral budding [18]. Further studies of retroviral budding by the Rous sarcoma virus (RSV) and the equine infectious anemia virus (EIAV) led to the discovery of other late domain motifs including PPXY and YPX(n)L, which also mimic motifs present in ESCRT components. These late domain motifs initiate assembly of the machinery through the binding of the E3-ubiquitin ligase NEDD4 and the ESCRT adaptor protein ALIX, respectively [1925]. Numerous mammalian viruses have proteins that encode late domain motifs that function as adaptors for the host ESCRT machinery. Intriguingly, viruses can encode multiple late domain motifs within the same adaptor protein, providing evidence for the importance of exploiting this host machinery for their pathogenesis. The newly described sequence motifs PLPPV and FPIV have been shown to also provide late domain activity for viral budding of mouse mammary tumor virus (MMTV) and paramyxoviruses, respectively [2629]. Although the ESCRT proteins recruited through the PLPPV motif remain to be identified, the FPIV motif encoded in Newcastle disease virus matrix protein (M protein) appears to facilitate an interaction with CHMP4B [30]. Nonetheless, it is well established that viral late domain motifs have evolved to mimic the function of proline-rich motifs that facilitate physiological and structural organization of ESCRT components [8]. Table 2 provides an overview of the mechanisms for microbial strategies for usurping of the host ESCRTs.

thumbnail
Table 2. Strategies of microbial pathogens in exploiting the host ESCRT machinery.

https://doi.org/10.1371/journal.ppat.1011344.t002

ESCRT homologues in prokaryotic cells and their contributions to phage pathogenesis

Fascinatingly, ESCRT-III and VPS4A homologs can be found in archaea, and there is evidence for the exploitation of these factors by an archaeal virus. In the case of prokaryotes, the ESCRT machinery is significantly reduced. The few homologues identified in Sulfolobus spp. correspond to the late acting ESCRT components from ESCRT-III and VPS4 [99,100]. These plant ESCRT homologs are exploited by the archaeal Sulfolobus turreted icosahedral virus (STIV) for replication [31]. Although a specific role for the ESCRT-III homologs has not been defined, a similar observation of ring-like structures at budding sites has been described for the archaeal Sulfolobus spindle-shaped virus-1 (SSV1) [101].

Homologues of ESCRT components have also been identified in the genomes of Asgard archaea [102]. Genomic analysis of these archaea have identified multiple genes that were thought to be unique to eukaryotes [103]. This novel finding is consistent with Asgard archaea and eukaryotes sharing a common ancestry. Among the eukaryotic signature proteins present in the Asgard archaea superphylum, several are associated with vesicular trafficking, including ESCRT-I-, ESCRT-II-, ESCRT-III- and ubiquitin modifier-like proteins [103]. Functional analysis of these proteins showed that the VPS4A homologue can bind ESCRT-III-like proteins and partially complement the loss of VPS4A in Saccharomyces cerevisiae [102]. This has led to recognizing the importance of this ancient machinery in evolutionary distinct cells.

Plant pathogens and their interactions with their host ESCRT

Although plants lack orthologs of ESCRT-0, they have components of ESCRT-I, -II, and -III that function in endosomal sorting (i.e., MVB biogenesis) and nonendosomal sorting events [104], including exploitation by pathogens (Fig 1). The plant virus tomato bushy stunt virus (TBSV) forms viral replication organelles in peroxisomes that are composed of vesicle-like structures termed spherules or viral replication complexes (VRCs), which are coated with the viral replicase protein p33 [105108]. These structures have been reproduced in yeast to study host factors necessary for their biogenesis [109,110]. A genome-wide screen using this model host identified several ESCRT components that are necessary for TBSV replication [111]. During the formation of VRCs at the peroxisome membrane, p33 is ubiquitinated and recruits the ESCRT-I component Vps23 (TSG101 homologue) through a late domain motif-like sequence, PSVP [112]. Additionally, the ESCRT accessory protein Bro1 (a homologue of human ALIX), the ESCRT-III components Vps24 and Snf7 (homologs of human CHMP3 and CHMP4, respectively), and Vps4 are important for VRC assembly during TBSV infection [32,34]. Surprisingly, host Vps4 appears to remain at the sites of VRCs following recruitment [33], differently from its canonical role in transiently localizing to the site of ESCRT complex assembly to remodel and disassemble the machinery [113]. As a result, the VRC remains “opened” to the host cytosol, and this configuration is also dependent on the function of ESCRT-III components at this structure where the viral RNA is protected from clearance by host intrinsic defenses [33,34].

thumbnail
Fig 1. The role of plant ESCRTs in microbial infection.

Plant viruses make use of the host ESCRT machinery for the formation of VRCs at different host organelles: (1) TBSV at peroxisomes via the viral protein p33 encoding a P[S/T]AP-like late domain motif; (2) BMV at the mitochondria via the viral protein 1a N-terminus; and (3) CIRV at the ER. (4) Plant ESCRTs also contribute to the formation of EVs that are secreted in response to pathogens as a mechanism for intrinsic defense. Created using Biorender.com. BMV, brome mosaic virus; CIRV, carnation Italian ringspot virus; ER, endoplasmic reticulum; ESCRT, Endosomal Sorting Complex Required for Transport; EV, extracellular vesicle; TBSV, Tomato bushy stunt virus; VRC, viral replication complex.

https://doi.org/10.1371/journal.ppat.1011344.g001

Other plant viruses that form ESCRT-dependent VRCs at the host mitochondria and endoplasmic reticulum (ER) are the carnation Italian ringspot virus (CIRV) [35] and the brome mosaic virus (BMV) [36,114], respectively. CIRV, a tombusvirus similar to TBSV, interacts with the host ESCRT-I component Vps23 through the viral protein p36 [35]. However, the CIRV p36 protein interacts with Vps23 by “mimicking” the Vps23–Vps28 interaction through the Vps23 StBox (steadiness box) domain [35]. This is different from the TBSV p33 protein and those of other viruses that interact with Vps23 UEV (ubiquitin E2 variant) domain through a P[S/T]AP-like motif [13,18,112]. In the case of BMV, several ESCRT components colocalize with viral protein 1a and are important for the replication of the virus, with host Snf7 (a member of ESCRT-III) depletion having the most significant effect in VRC formation [36].

Another relevant aspect of the ESCRT machinery in plants is its function in forming extracellular vesicles (EVs) and their role in the defense against microbes and facilitating plant–microbe symbiosis, as recently reviewed by Cui and colleagues [115]. Fusion of MVBs with the plasma membrane results in the release of EVs that function in immune defense against plant pathogens [116]. Secreted EVs contain proteins that stimulate fortification of the cell wall as a barrier against pathogens [115]. Plant EVs also contain small RNAs that can target fungal pathogens to induce gene silencing [117]. As a similar mechanism of defense, EV secretion appears to be increased upon Pseudomonas syringae infection [118]. However, the role of individual ESCRT components in MVB formation for EV-mediated pathogen defense, and the extent to which this is conserved in mammals, remains to be elucidate.

Microbial strategies for usurping host ESCRTs in animal cells

ESCRT-dependent envelopment: Viral budding at the host plasma membrane.

The budding of enveloped viruses has parallels to ESCRT-dependent MVB formation (Fig 2). The role of ESCRTs in this process has been extensively reviewed [119122]. In some cases, ubiquitination is an important factor for the recruitment of the ESCRTs for viral budding. This is mainly the case for viruses encoding the PPXY late domain motif for the recruitment of the E3-ubiquitin ligase NEDD4, including EBOV, RSV, human T-cell leukemia virus-1 (HTLV-1), murine leukemia virus (MLV), and Marburg virus (MARV) [122]. Ubiquitinated cargo can be recognized by several ESCRT components including the ESCRT-0 HRS protein, the ESCRT-I components TSG101 and Vps23, and the ESCRT accessory protein ALIX [119]. As such, viruses that use the PPXY-NEDD4-ubiquitination strategy have effectively evolved an alternative mechanism for interacting with TSG101 and ALIX in the absence of P[S/T]AP and YPX(n)L late domain motifs [123].

thumbnail
Fig 2. Mechanisms of ESCRT exploitation by RNA viruses.

(1) Retroviruses, such as the HIV-1, interact with the host ESCRT machinery for viral budding via structural proteins like Gag, encoding the P[S/T]AP and YPXL late domain motifs. Other RNA viruses like (2) DENV and (3) HCV recruit host ESCRT components to the ER to promote viral replication. This recruitment is facilitated by nonstructural proteins (NS) via unknown mechanisms. Created using Biorender.com. DENV, dengue virus; ER, endoplasmic reticulum; ESCRT, Endosomal Sorting Complex Required for Transport; HCV, hepatitis C virus; HIV-1, human immunodeficiency virus-1.

https://doi.org/10.1371/journal.ppat.1011344.g002

Gag is the main structural protein for retrovirus assembly [124]. During HIV-1 release, which is the most studied retroviral ESCRT-mediated budding process, Gag localizes to the plasma membrane of host cells where it interacts with TSG101 through the PTAP motif encoded in its p6 domain [13,15,16,37]. Interestingly, the HIV-1 Gag PTAP peptide binds the host TSG101 UEV domain with a higher affinity than the HRS PTAP peptide [37]. Disrupting either the HIV-1 PTAP motif or the host TSG101 impairs virus release [14,15,17]. Additionally, HIV-1 Gag encodes a second late domain motif, YPX(n)L, which binds to the ALIX V domain [39]; however, its binding is weaker than that of the EIAV Gag YPX(n)L late domain motif [22]. Nonetheless, ALIX contributes in HIV-1 virus release, and overexpressing it can restore the budding deficiency of HIV-1 PTAP mutants [22,39,40]. ESCRT-III and VPS4 complexes are also critical for HIV-1 viral budding [15,125]. The role of ALIX in viral budding was confirmed using EIAV since this virus lacks a PTAP motif and solely relies on a YPX(n)L motif [41].

Recruitment dynamics of ESCRT components to HIV-1 viral assembly sites at the plasma membrane have supported a role for TSG101, ALIX, CHMP4B, and VPS4. Studies using superresolution microscopy provided evidence for the colocalization of TSG101, ALIX, and CHMP4B to the cell membrane where HIV-1 puncta accumulate [126]. These observations were reinforced by live imaging studies [127129]. Such studies support a model in which HIV-1 Gag recruits TSG101 to the cell plasma membrane where it remains at the site of virus-like particle (VLP) budding, while the late acting ESCRT proteins CHMP4B and VPS4A are transiently recruited to these sites. The recruitment of these late acting ESCRT components is not simultaneous; CHMP4B localized to the nascent VLP a few seconds before VPS4A arrived, but CHMP4B departed within approximately 18 s, whereas VPS4A lingered for approximately 48 s [128,130]. CHMP4B and ALIX assemble simultaneously and their disassembly is dependent on the recruitment and ATPase function of VPS4 [129].

Overall, the viral requirements for early ESCRT components (ESCRT-I, ALIX) appear to vary according to their late domain motifs; however, they converge in the recruitment of CHMP4 and VPS4 isoforms to mediate membrane fission and release of viral particles [41,119].

Strategies used by other RNA viruses: Viral particle assembly at intracellular compartments.

Although the best described examples of a pathogen usurping the ESCRT machinery are seen in viral egress from the host cell, other RNA viruses have developed their own strategies for manipulating this multifunctional host machinery (Fig 2). Different from ESCRT-dependent viral budding, these viruses do not interact with host ESCRTs through their structural proteins. As previously described for plant viruses, animal cell viruses from the Flaviviridae family also form cytoplasmic VRCs [131], and their viral nonstructural proteins (NS) are key components for the formation of these important replicative sites [132]. This group of viruses, which includes dengue virus (DENV), Japanese encephalitis virus (JEV), and yellow fever virus (YFV), replicate at VRCs comprised of ER-derived membranous compartments [133].

The role of host ESCRT machinery in Flaviviridae pathogenesis remains poorly understood compared to viral budding. However, due to the similarities of their replicative complexes to plant viruses, studies have focused on elucidating the potential role of host ESCRTs in this process. Even though late domain motifs had not been identified in Flaviviruses until recently [61], it was known that TSG101 and ALIX can interact with the nonstructural protein NS3 expressed by JEV, DENV, and YFV [51,52,54]. Knocking down TSG101, CHMP2, or CHMP4 reduced JEV and DEV viral titers, and ESCRT-III depletion also affected the number of mature virions particles present at VRCs [53]. Additionally, the protein levels of host ALIX significantly influenced the outcome of viral particles titers in the case of DENV infection [54]. Host CHMP2 and CHMP4 also localized to JEV VRCs, suggesting that they promote membrane deformation for viral particle formation [53].

A more comprehensive depiction of the mechanism for ESCRT-dependent VRC formation was provided in a recent study of classical swine fever virus (CSFV) infection [60]. Using an siRNA screen, the authors identified several ESCRT components that contribute to CSFV infection at different stages of infectious cycle, including entry. Comparable to what has been observed for JEV, the ESCRT-III components CHMP2 and CHMP4 localized to CSFV VRCs [53,60]. Although ESCRT components such as HRS, TSG101, VPS28 (ESCRT-I), EAP20 (ESCRT-II), ALIX, CHMP7, and VPS4 also contribute to VRC formation through their interactions with the nonstructural proteins NS3, NS4B, NS5A, and NS5B [60], the molecular mechanism by which these nonstructural proteins interact with host ESCRT components remains to be elucidated.

Another important human virus within the Flaviviridae family is the hepatitis C virus (HCV). Differently from DENV virus, which forms single membrane VRCs, HCV forms ER-derived VRCs composed of double membrane vesicles [134]. Interestingly, HCV also exploits host ESCRTs during viral particle formation [55,58]. In this case, host ESCRTs might be recruited to the site of virus particle assembly through the nonstructural HCV protein NS2 that can interact with the ubiquitin interaction motif domain of the ESCRT-0 component HRS [56]. Since no canonical late domain motifs have been identified in Flaviviridae nonstructural proteins discussed here, recruitment of host ESCRTs for VRC formation presumably occurs through novel motifs.

DNA viruses: Exploiting the host ESCRT to exit the nucleus.

DNA viruses also make use of the host ESCRT machinery at different steps of their infectious cycle (Fig 3). In the case of Herpesviridae family viruses, there is emerging evidence for a role of host ESCRTs in primary envelopment of herpes simplex virus-1 (HSV-1) and Epstein–Barr virus (EBV) at the inner nuclear membrane (INM) [69,70,76]. There are 2 main proteins in herpesviruses that form the viral nuclear egress complex (NEC), namely UL31 and UL34 in HSV-1, and BFLF2 and BFRF1 in EBV [135]. NECs function in primary envelopment by interacting with the DNA-containing viral nucleocapsid to facilitate budding of enveloped virion from the INM to the perinuclear space [135]. HSV-1 uses UL31 and UL34 to recruit CHMP4 and ALIX to the INM [69]. Knockdown of CHMP4 or ALIX led to the accumulation of virions at the nucleus, providing evidence for a role of the host ESCRT machinery in HSV-1 primary envelopment [69]. Recent work showed that UL34 interacts with ALIX though a novel mechanism involving arginine clusters at its C-terminal [70]. Although these arginine clusters are not conserved in the EBV UL34 homologue BFRF1 [70], a role for BFRF1 in the recruitment of CHMP4 and ALIX has also been reported [76]. BFRF1 recruitment of ESCRT components could be through its ubiquitination, which has been demonstrated to contribute to NEC formation [77]. Although ESCRTs play a basic role in maintaining the nuclear envelope [2], the topology for its proposed role in Herpesviridae primary envelopment at the INM (i.e., into the cytosol) is opposite of how it typically functions (i.e., away from the cytosol).

thumbnail
Fig 3. Mechanisms of ESCRT exploitation by DNA viruses.

(1) The DNA virus HSV-1 requires ESCRT components for (1a) primary envelopment at the nucleus via the arginine-rich cluster at the UL34 viral protein and (1b) secondary envelopment at the trans-Golgi via the P[S/T]AP and YPXL late domain motifs encoded by UL36. (2) VACV recruits the ESCRT machinery for viral envelopment and egress from MVBs. The viral protein F13L has been proposed to interact with ESCRT components via YPXL late domain motifs. Created using Biorender.com. ESCRT, Endosomal Sorting Complex Required for Transport; HSV-1, herpes simplex virus-1; MVB, multivesicular body; VACV, vaccinia virus.

https://doi.org/10.1371/journal.ppat.1011344.g003

Following HSV-1 capsid release into the host cytoplasm via the fusion of the INM-derived enveloped virion with the outer nuclear membrane (ONM) [135], a second envelopment step occurs at the host trans-Golgi [136,137]. Host ESCRT also contributes at this step, as observed by the accumulation of partially enveloped viral particles upon disrupting this machinery using a VPS4 dominant negative (VPS4EQ) form [72,73]. The viral protein UL36 (also known as VP1/2), which interacts with TSG101 [74], could be partially responsible for the recruitment of host ESCRTs for secondary envelopment even though depletion of TSG101 does not seem to impair HSV-1 production [73,75]. Remarkably, the HSV-1 viral protein UL51 has structural homology to CHMP4B and is capable of forming ESCRT-III-like filaments [138]. It would be interesting to determine if ESCRT-III components recognize and interact with UL51 filaments to promote viral assembly and the extent to which VPS4 functions in their disassembly.

Interactions of intracellular bacteria with host ESCRT.

Most intracellular bacteria reside within a membrane-bound compartment (the pathogen-containing vacuole or PCV) that is modified by the pathogen to promote its survival. Emerging evidence demonstrates that the host ESCRT machinery could also be targeted by intracellular bacteria residing in a PCV (Fig 4). For example, the obligate intracellular bacteria Anaplasma phagocytophilum resides within a host cell–derived vacuole that receives membrane traffic from multiple sources, likely to satisfy its metabolic needs [139]. A recent study showed that the A. phagocytophilum (PCV) resembles an MVB that is decorated with ALIX and ESCRT-0 and ESCRT-III components. Remarkably, knocking down ALIX or components of ESCRT-0 or ESCRT-III not only arrested bacterial growth but also prevented release of progeny by blocking fusion of the PCV with the plasma membrane, which is akin to MVB release of exosomes [86]. This study suggests that A. phagocytophilum benefits from residing in an MVB-like compartment through the ESCRT-dependent delivery of material and ESCRT-mediated release of infectious progeny. The release of bacteria encased in MVBs has also been previously reported for Brucella abortus and uropathogenic Escherichia coli (UPEC) [87,88]. It remains to be determined which bacteria effector proteins promote formation of the MVB-like compartment enclosing the bacteria. ESCRT function seems to promote A. phagocytophilum and B. abortus progeny release [86,87]; however, in the context of UPEC infection, it has been proposed to be a defense mechanism against the pathogen [88].

thumbnail
Fig 4. The host ESCRT machinery and nonviral pathogens.

The outcome of intracellular bacterial infection is affected by the host ESCRT machinery. (1) Brucella abortus recruits ESCRT components to its PVC, which resembles an MVB, to promote proliferation and release. (2) Salmonella spp. recruit ESCRT components for the formation of its PCV. The bacterial protein SpoB has been proposed to mediate this interaction. (3) Differently from A. phagocytophilum and Salmonella, Mycobacterium tuberculosis inhibits the function of the host ESCRT machinery to evade clearance and promote its survival. The bacteria secrete the effector proteins EsxG and EsxH to inhibit ESCRT-dependent PCV repair (4) The UPEC are encased in MVBs by the host ESCRT machinery for expulsion from cells, a mechanism for ESCRT-mediated intrinsic defense. Created using Biorender.com. ESCRT, Endosomal Sorting Complex Required for Transport; MVB, multivesicular body; PVC, pathogen-containing vacuole; UPEC, uropathogenic Escherichia coli.

https://doi.org/10.1371/journal.ppat.1011344.g004

Other intracellular bacteria including Salmonella spp., Coxiella burnetii, and Mycobacterium tuberculosis have developed mechanisms to avoid lysosomal dependent clearance and reside in PCVs that promote bacterial persistence and proliferation [140,141]. Salmonella can form a PCV in nonphagocytic cells, and a role for host ESCRTs in the biogenesis of its PVC has been recently proposed [91,92]. ESCRT-III components localized to Salmonella PCVs, and bacteria invading CHMP3 knockout cells were exposed to the cytosol due to the formation of aberrant PCVs [91]. The ESCRT-III component CHMP4B was also reported to be recruited to the C. burnetii PCV [94]. In this case, ESCRT recruitment was associated with PCV damage as determined by the co-recruitment of Galectin-3 [94]. CHMP4B and Galectin-3 recruitment to C. burnetii PCV was transient, potentially corresponding to membrane repair and promoting bacterial survival; this is consistent with the finding that TSG101 depletion reduced bacterial replication [94]. These studies suggest a beneficial role for host ESCRTs in supporting Salmonella’s and C. burnetii’s intracellular niche by contributing to the integrity of the PCV. Conversely, studies of M. tuberculosis demonstrated that knockdown of ESCRT components increased bacterial growth [89,142], suggesting an antagonistic role for host ESCRT. Additional work showed that host ESCRT machinery is recruited to the mycobacterial PCV in response to membrane damage and that membrane repair promotes lysosomal maturation leading to bacterial clearance [90]. As a mechanism to avoid clearance, mycobacteria effector proteins EsxG and EsxH disrupt host ESCRTs to promote bacterial survival [89,90].

An intracellular parasite’s approach for exploiting the host ESCRT machinery.

Although decades of research described above have identified many different intracellular pathogens that interact with host ESCRTs, a role for ESCRTs during replication of an intracellular eukaryotic microorganism has only recently emerged (Fig 5). Toxoplasma gondii is an intracellular parasite capable of manipulating the host cell with secretory effector proteins to promote its intracellular survival. One class of effector proteins (GRAs) are secreted from dense granule organelles after the parasite has invaded a cell to create its intracellular niche, the parasitophorous vacuole (PV) [143]. A subset of GRA proteins integrate into the PV membrane with a single transmembrane segment, thereby bridging the PV lumen and host cytosol [144]. Residing at this parasite–host interface, such proteins are ideally positioned to communicate with the infected cell including for the acquisition of resources [145]. T. gondii acquires proteins and lipids from infected cells by vesicular uptake or entrapment, respectively, within the PV [146,147]. A potential role for the host ESCRT machinery for these processes was hypothesized since it would involve budding of vesicles away from the host cytosol and into the lumen of the PV, consistent with ESCRT function. Accordingly, a recent study identified several ESCRT proteins associated with the PV based on proximity labelling experiments [148]. Additional work showed that ESCRT-III accumulates at the PV upon overexpression of dominant negative VPS4A [95]. Bioinformatic searches identified a PV membrane resident secretory protein called GRA14, which encodes several ESCRT late domain motifs in its C-terminus that are exposed to the host cytosol. Further studies established that GRA14 interacts with the host ESCRT machinery through its late domain motifs to mediate the vesicular uptake of host cytosolic proteins across the PV membrane [95]. Interestingly, recruitment of TSG101, but not ALIX, is dependent on GRA14, suggesting the contribution of other parasite effector proteins in the recruitment of the multiple ESCRT components observed at the PV [95]. The transmembrane dense granule protein GRA64 is a candidate for ALIX recruitment since it was shown to co-immunoprecipitate with ESCRT components [96]. Although GRA64 lacks apparent late domain motifs, it has an arginine cluster like that of the HSV-1 UL34 protein [96]. Moreover, Romano and colleagues [97] recently reported that parasites lacking GRA64 or GRA14 entrap fewer intra-PV host organelles. Disrupting both GRA64 and GRA14 resulted in an additive effect, suggesting that these proteins have nonredundant functions. This study also showed that CHMP4B is recruited to the PV and that CHMP4B forms striking spiral filament within PV membrane invaginations when expressed as a dominant negative mutant. The authors further report that expression of dominant negative VPS4A caused the accumulation of entrapped host-derived endolysosomal vesicles in the PV. Together, these studies suggest that T. gondii uses multiple effector proteins to exploit host ESCRTs for vesicular uptake or PV entrapment of host-derived resources.

thumbnail
Fig 5. gondii has different strategies for exploiting the host ESCRT machinery.

T. (1) The protozoan parasite Toxoplasma gondii recruits the early ESCRT components TSG101 and ALIX during invasion via the P[S/T]AP and YPXL late domain motifs encoded by RON5 and RON2, respectively. (2) While residing in its replicative compartment known as the PV, the parasite recruits ESCRT components for the uptake of resources from the host cell across the PV membrane. (2a) GRA14 encodes both P[S/T]AP and YPXL late domain motifs and is required for the recruitment of the host ESCRT machinery for the uptake of host cytosolic proteins. (2b) Additionally, GRA14 alongside another parasite ESCRT-interacting protein GRA64, are necessary for the sequestration of host vesicles. Created using Biorender.com. ESCRT, Endosomal Sorting Complex Required for Transport; PV, parasitophorous vacuole.

https://doi.org/10.1371/journal.ppat.1011344.g005

T. gondii also interacts with TSG101 and ALIX during parasite invasion into host cells, in this case using RON proteins derived from the necks of rhoptry secretory organelles [98]. The role of TSG101 and ALIX in parasite invasion, however, does not appear to be linked to their function in endosomal sorting since other components of ESCRT-I, ESCRT-III, or VPS4 were absent at the site of parasite invasion [98]. Thus, T. gondii appears to have multiple strategies for exploiting the host ESCRT machinery at different steps during infection, serving as an interesting model for studying these microbe–host interactions.

Could there be a role for the ESCRT machinery in immunity?

A significant discovery for the multiple functions of the ESCRT has been its role in repairing membrane damage to protect cancer cells targeted by cytotoxic T cells [149]. The ESCRT machinery can negatively regulate apoptosis, necroptosis, pyroptosis, and ferroptosis to promote cell survival through different mechanisms [150]. For example, disruption of ESCRT function increased inflammasome activation in response to pathogen-associated molecular patterns (PAMPs), leading to pyroptotic cell death [151]. This is due to the anti-inflammatory role of the ESCRT machinery in repairing gasdermin D (GSDMD) pores at the host plasma membrane [151]. Since some of these cell death pathways are activated in infected cells to limit pathogen replication, pathogens could modulate host ESCRTs as a potential immune evasion strategy. Conversely, ESCRT function in membrane repair could be a mechanism for immune cell protection from microbial pore forming proteins that cause membrane lesions. An example of the latter has been reported in the context of Candida albicans infection [152]. To protect themselves from lesions caused by the fungal toxin candidalysin, epithelial cells dispose of damaged membrane in an ALG-2/ESCRT-dependent manner [152]. This resembles the contribution of the ESCRT machinery in plants for the protection against pathogens.

Although these examples correspond to the membrane repair function of the ESCRT machinery, its canonical role in vesicular trafficking could also impact the function of signalling pathways [153]. For example, the ESCRT machinery can terminate STING (stimulator of interferon genes) signalling, an important modulator of the type I interferon immune response [154]. Additionally, HRS, STAM2, TSG101, and VPS4 are important for sorting of ubiquitinated T cell receptor into microvesicles for signal termination [155157]. The ESCRT machinery also negatively regulates antigen cross-presentation in dendritic cells by repairing phagosomal membrane damage, interrupting the export of antigens to the cytosol [158]. A recent report showed that dendritic cells infected with T. gondii up-regulate the expression of the ESCRT-III component CHMP4B and that CHMP4B is associated with the PV [159]. The same study showed that blocking cholesterol trafficking impaired MHC-I and MHC-II presentation of parasite antigens, reduced PV recruitment of CHMP4B, and suppressed parasite replication [159]. Although the role of the ESCRT machinery in linking these phenotypes hasn’t been elucidated, it would be interesting to know if up-regulation of ESCRT components negatively regulates antigen presentation.

Concluding remarks

Evidently, microbial exploitation of the host ESCRT machinery is not limited to viral budding or viruses altogether. Whereas viruses appear to benefit mostly from the vesicular budding features of the host ESCRT machinery, bacteria make use of the membrane remodelling function of the host ESCRT machinery during infection [160]. Future studies should focus on how pathogens recruit the host ESCRT machinery to maintain an intact PCV, as for C. burnetii infection [94], or promoting the formation of MVB-like compartments encapsulating the bacteria [86,87]. Also, how could this be controlled to avoid expulsion from the cells as occurs for UPEC infection [88,161]? What are the factors dictating the different outcomes?

Another interesting discovery to the field is that a eukaryotic pathogen can manipulate the host ESCRT machinery. Protozoan parasites encode a reduced ESCRT machinery that is still functional in EV biogenesis [162,163]. Why does T. gondii, encoding ESCRT-homologs and capable of producing EVs, prioritize the use of the host ESCRT machinery when the related parasite Plasmodium falciparum secretes its ESCRT homologs in to the host cytosol to promote EV formation [164]? Furthermore, T. gondii can exploit the host ESCRT machinery for invasion and at least 2 different pathways for nutrient acquisition, could it also be making use of the ESCRT machinery for other purposes? To our knowledge, this is the only microorganism capable of exploiting the host ESCRT machinery for multiple cellular functions. Are there other intracellular protozoan parasites capable of interacting with the host ESCRT machinery or do they rely on their own ESCRT machinery?

The apparent role for the ESCRT machinery in immunity opens the question as to whether pathogens modulate the expression of ESCRT components to influence the outcome of infection. The ESCRT machinery can terminate STING signalling; however, it is also necessary for sorting of foreign DNA into EVs to stimulate STING signalling in bystander cells [154,165]. STING signalling promotes inflammasome activation, which results in the assembly of GSDMD pores at the plasma membrane leading to pyroptotic cell death, a process that is tempered by the ESCRT machinery [151]. Thus, much remains to be understood for the function of the ESCRT machinery in innate immunity, the extent to which this is regulated, and how microbes could be exploiting this.

Fundamental knowledge of ESCRT biology has been gained through the study of this host machinery during viral infection. Most notably, identifying the P[S/T]AP late domain motif in HIV-1 Gag and its interaction with TSG101 preceded the realization that these motif were present in HRS and ALIX and that retroviruses were “mimicking” intercomplex interactions to make use of the host ESCRT machinery [13,166]. With the many other pathogen–ESCRT interactions that been identified but not mechanistically understood (Fig 6), a treasure trove of insight is likely yet to be gained about how pathogens exploit ESCRTs, thereby exposing new basic mechanisms of this remarkably versatile membrane remodelling machinery.

thumbnail
Fig 6. A pathogen’s guide for exploiting the host ESCRT machinery.

ESCRT are involved in multiple functions in cells and many intracellular pathogens have evolved ways to exploit ESCRTs for key aspects of their pathogenesis. Viral envelopment and budding: For viral envelopment and budding, viruses encode short linear amino acid motifs that mimic ESCRT interactions necessary for ESCRTs sequentially assemble. Viral replication: Alternatively, viruses can recruit host ESCRT components for the formation of replication complexes at host organelles. These strategies used by RNA and DNA viruses resemble MVB formation by ESCRT. Bacterial survival, maintenance of replicative compartment, and release: Bacteria can also benefit from the host ESCRT machinery for the biogenesis, maintenance (ESCRT membrane repair function), and release of their replicative compartment (ESCRT MVB formation and exosome release). Parasite invasion and resource acquisition: The protozoan parasite Toxoplasma gondii recruits early ESCRT components for invasion and subsequently during replication for resource acquisition. Intrinsic defense: Interestingly, the role of ESCRT has also been associated with intrinsic defenses against pathogens, for example, expulsion of infecting bacteria and enclosing foreign DNA in exosomes for immune signalling. Created using Biorender.com. ESCRT, Endosomal Sorting Complex Required for Transport; MVB, multivesicular body.

https://doi.org/10.1371/journal.ppat.1011344.g006

Acknowledgments

We thank members of our lab and the labs of Isabelle Coppens and Louis Weiss for helpful discussions.

References

  1. 1. Hurley JH. ESCRTs are everywhere. EMBO J. 2015 Oct 1;34(19):2398–2407. pmid:26311197
  2. 2. Vietri M, Radulovic M, Stenmark H. The many functions of ESCRTs. Nat Rev Mol Cell Biol. 2020 Jan;21(1):25–42. pmid:31705132
  3. 3. Isono E. ESCRT Is a Great Sealer: Non-Endosomal Function of the ESCRT Machinery in Membrane Repair and Autophagy. Plant Cell Physiol. 2021 Oct 1;62(5):766–774. pmid:33768242
  4. 4. Campsteijn C, Vietri M, Stenmark H. Novel ESCRT functions in cell biology: spiraling out of control? Curr Opin Cell Biol. 2016 Aug;41:1–8. pmid:27031044
  5. 5. Schmidt O, Teis D. The ESCRT machinery. Curr Biol. 2012 Feb 21;22(4):R116–R120. pmid:22361144
  6. 6. Adell MAY, Migliano SM, Teis D. ESCRT-III and Vps4: a dynamic multipurpose tool for membrane budding and scission. FEBS J. 2016;283(18):3288–3302. pmid:26910595
  7. 7. Wollert T, Yang D, Ren X, Lee HH, Im YJ, Hurley JH. The ESCRT machinery at a glance. J Cell Sci. 2009 Jul 1;122(Pt 13):2163–2166. pmid:19535731
  8. 8. Ren X, Hurley JH. Proline-Rich Regions and Motifs in Trafficking: From ESCRT Interaction to Viral Exploitation. Traffic. 2011;12(10):1282–1290. pmid:21518163
  9. 9. Henne WM, Buchkovich NJ, Emr SD. The ESCRT Pathway. Dev Cell. 2011 Jul 19;21(1):77–91.
  10. 10. Alonso Y, Adell M, Teis D. Assembly and disassembly of the ESCRT-III membrane scission complex. FEBS Lett. 2011;585(20):3191–3196. pmid:21924267
  11. 11. von Schwedler UK, Stuchell M, Müller B, Ward DM, Chung HY, Morita E, et al. The protein network of HIV budding. Cell. 2003 Sep 19;114(6):701–713. pmid:14505570
  12. 12. Meng B, Lever AM. Wrapping up the bad news–HIV assembly and release. Retrovirology. 2013 Jan 10;10:5. pmid:23305486
  13. 13. Pornillos O, Higginson DS, Stray KM, Fisher RD, Garrus JE, Payne M, et al. HIV Gag mimics the Tsg101-recruiting activity of the human Hrs protein. J Cell Biol. 2003 Aug 4;162(3):425–434. pmid:12900394
  14. 14. Huang M, Orenstein JM, Martin MA, Freed EO. p6Gag is required for particle production from full-length human immunodeficiency virus type 1 molecular clones expressing protease. J Virol. 1995 Nov;69(11):6810–6818. pmid:7474093
  15. 15. Garrus JE, von Schwedler UK, Pornillos OW, Morham SG, Zavitz KH, Wang HE, et al. Tsg101 and the vacuolar protein sorting pathway are essential for HIV-1 budding. Cell. 2001 Oct 5;107(1):55–65. pmid:11595185
  16. 16. VerPlank L, Bouamr F, LaGrassa TJ, Agresta B, Kikonyogo A, Leis J, et al. Tsg101, a homologue of ubiquitin-conjugating (E2) enzymes, binds the L domain in HIV type 1 Pr55Gag. Proc Natl Acad Sci. 2001 Jul 3;98(14):7724–7729. pmid:11427703
  17. 17. Göttlinger HG, Dorfman T, Sodroski JG, Haseltine WA. Effect of mutations affecting the p6 gag protein on human immunodeficiency virus particle release. Proc Natl Acad Sci U S A. 1991 Apr 15;88(8):3195–3199. pmid:2014240
  18. 18. Licata JM, Simpson-Holley M, Wright NT, Han Z, Paragas J, Harty RN. Overlapping motifs (PTAP and PPEY) within the Ebola virus VP40 protein function independently as late budding domains: involvement of host proteins TSG101 and VPS-4. J Virol. 2003 Feb;77(3):1812–1819. pmid:12525615
  19. 19. Parent LJ, Bennett RP, Craven RC, Nelle TD, Krishna NK, Bowzard JB, et al. Positionally independent and exchangeable late budding functions of the Rous sarcoma virus and human immunodeficiency virus Gag proteins. J Virol. 1995 Sep;69(9):5455–5460. pmid:7636991
  20. 20. Puffer BA, Parent LJ, Wills JW, Montelaro RC. Equine infectious anemia virus utilizes a YXXL motif within the late assembly domain of the Gag p9 protein. J Virol. 1997 Sep;71(9):6541–6546. pmid:9261374
  21. 21. Strack B, Calistri A, Accola MA, Palù G, Göttlinger HG. A role for ubiquitin ligase recruitment in retrovirus release. Proc Natl Acad Sci U S A. 2000 Nov 21;97(24):13063–13068. pmid:11087860
  22. 22. Fisher RD, Chung HY, Zhai Q, Robinson H, Sundquist WI, Hill CP. Structural and biochemical studies of ALIX/AIP1 and its role in retrovirus budding. Cell. 2007 Mar 9;128(5):841–852. pmid:17350572
  23. 23. Garnier L, Wills JW, Verderame MF, Sudol M. WW domains and retrovirus budding. Nature. 1996 Jun 27;381(6585):744–745. pmid:8657277
  24. 24. Yasuda J, Hunter E, Nakao M, Shida H. Functional involvement of a novel Nedd4-like ubiquitin ligase on retrovirus budding. EMBO Rep. 2002 Jul;3(7):636–640. pmid:12101095
  25. 25. Iglesias-Bexiga M, Palencia A, Corbi-Verge C, Martin-Malpartida P, Blanco FJ, Macias MJ, et al. Binding site plasticity in viral PPxY Late domain recognition by the third WW domain of human NEDD4. Sci Rep. 2019 Oct 21;9(1):15076. pmid:31636332
  26. 26. Coren LV, Nagashima K, Ott DE. A PLPPV Sequence in the p8 Region of Gag Provides Late Domain Function for Mouse Mammary Tumor Virus. Virology. 2019 Sep;535:272–278. pmid:31357166
  27. 27. Schmitt AP, Leser GP, Morita E, Sundquist WI, Lamb RA. Evidence for a New Viral Late-Domain Core Sequence, FPIV, Necessary for Budding of a Paramyxovirus. J Virol. 2005 Mar;79(5):2988–2997. pmid:15709019
  28. 28. Li M, Schmitt PT, Li Z, McCrory TS, He B, Schmitt AP. Mumps Virus Matrix, Fusion, and Nucleocapsid Proteins Cooperate for Efficient Production of Virus-Like Particles. J Virol [Internet]. 2009 Jul [cited 2022 Feb 4]; Available from: pmid:19439476
  29. 29. Duan Z, Hu Z, Zhu J, Xu H, Chen J, Liu H, et al. Mutations in the FPIV motif of Newcastle disease virus matrix protein attenuate virus replication and reduce virus budding. Arch Virol. 2014 Jul;159(7):1813–1819. pmid:24477785
  30. 30. Pei Y, Xue J, Teng Q, Feng D, Huang M, Liang R, et al. Mutation of Phenylalanine 23 of Newcastle Disease Virus Matrix Protein Inhibits Virus Release by Disrupting the Interaction between the FPIV L-Domain and Charged Multivesicular Body Protein 4B. Microbiol Spectr. 2023 Feb 14;11(1):e0411622. pmid:36695580
  31. 31. Snyder JC, Samson RY, Brumfield SK, Bell SD, Young MJ. Functional interplay between a virus and the ESCRT machinery in Archaea. Proc Natl Acad Sci. 2013 Jun 25;110(26):10783–10787. pmid:23754419
  32. 32. Barajas D, Jiang Y, Nagy PD. A Unique Role for the Host ESCRT Proteins in Replication of Tomato bushy stunt virus. PLoS Pathog. 2009 Dec 24;5(12):e1000705. pmid:20041173
  33. 33. Barajas D, Martín IF de C, Pogany J, Risco C, Nagy PD. Noncanonical role for the host Vps4 AAA+ ATPase ESCRT protein in the formation of Tomato bushy stunt virus replicase. PLoS Pathog. 2014 Apr;10(4):e1004087. pmid:24763736
  34. 34. Kovalev N, de Castro Martín IF, Pogany J, Barajas D, Pathak K, Risco C, et al. Role of Viral RNA and Co-opted Cellular ESCRT-I and ESCRT-III Factors in Formation of Tombusvirus Spherules Harboring the Tombusvirus Replicase. J Virol. 2016 Mar 11;90(7):3611–3626. pmid:26792735
  35. 35. Richardson LGL, Clendening EA, Sheen H, Gidda SK, White KA, Mullen RT. A unique N-terminal sequence in the Carnation Italian ringspot virus p36 replicase-associated protein interacts with the host cell ESCRT-I component Vps23. J Virol. 2014 Jun;88(11):6329–6344. pmid:24672030
  36. 36. Diaz A, Zhang J, Ollwerther A, Wang X, Ahlquist P. Host ESCRT Proteins Are Required for Bromovirus RNA Replication Compartment Assembly and Function. PLoS Pathog. 2015 Mar 6;11(3):e1004742. pmid:25748299
  37. 37. Im YJ, Kuo L, Ren X, Burgos PV, Zhao XZ, Liu F, et al. Crystallographic and Functional Analysis of the ESCRT-I /HIV-1 Gag PTAP Interaction. Struct Lond Engl 1993. 2010 Nov 10;18(11):1536–1547. pmid:21070952
  38. 38. Martin-Serrano J, Zang T, Bieniasz PD. HIV-1 and Ebola virus encode small peptide motifs that recruit Tsg101 to sites of particle assembly to facilitate egress. Nat Med. 2001 Dec;7(12):1313–1319. pmid:11726971
  39. 39. Strack B, Calistri A, Craig S, Popova E, Göttlinger HG. AIP1/ALIX Is a Binding Partner for HIV-1 p6 and EIAV p9 Functioning in Virus Budding. Cell. 2003 Sep 19;114(6):689–699. pmid:14505569
  40. 40. Usami Y, Popov S, Göttlinger HG. Potent Rescue of Human Immunodeficiency Virus Type 1 Late Domain Mutants by ALIX/AIP1 Depends on Its CHMP4 Binding Site. J Virol. 2007 Jun 15;81(12):6614–6622.
  41. 41. Sandrin V, Sundquist WI. ESCRT requirements for EIAV budding. Retrovirology. 2013 Oct 9;10(1):104. pmid:24107264
  42. 42. Wills JW, Cameron CE, Wilson CB, Xiang Y, Bennett RP, Leis J. An assembly domain of the Rous sarcoma virus Gag protein required late in budding. J Virol. 1994 Oct;68(10):6605–6618. pmid:8083996
  43. 43. Xiang Y, Cameron CE, Wills JW, Leis J. Fine mapping and characterization of the Rous sarcoma virus Pr76gag late assembly domain. J Virol. 1996 Aug;70(8):5695–5700. pmid:8764091
  44. 44. Kikonyogo A, Bouamr F, Vana ML, Xiang Y, Aiyar A, Carter C, et al. Proteins related to the Nedd4 family of ubiquitin protein ligases interact with the L domain of Rous sarcoma virus and are required for gag budding from cells. Proc Natl Acad Sci U S A. 2001 Sep 25;98(20):11199–11204. pmid:11562473
  45. 45. Vana ML, Tang Y, Chen A, Medina G, Carter C, Leis J. Role of Nedd4 and Ubiquitination of Rous Sarcoma Virus Gag in Budding of Virus-Like Particles from Cells. J Virol. 2004 Dec 15;78(24):13943–13953. pmid:15564502
  46. 46. Medina G, Zhang Y, Tang Y, Gottwein E, Vana ML, Bouamr F, et al. The functionally exchangeable L domains in RSV and HIV-1 Gag direct particle release through pathways linked by Tsg101. Traffic Cph Den. 2005 Oct;6(10):880–894. pmid:16138902
  47. 47. Harty RN, Brown ME, Wang G, Huibregtse J, Hayes FP. A PPxY motif within the VP40 protein of Ebola virus interacts physically and functionally with a ubiquitin ligase: implications for filovirus budding. Proc Natl Acad Sci U S A. 2000 Dec 5;97(25):13871–13876. pmid:11095724
  48. 48. Timmins J, Schoehn G, Ricard-Blum S, Scianimanico S, Vernet T, Ruigrok RWH, et al. Ebola virus matrix protein VP40 interaction with human cellular factors Tsg101 and Nedd4. J Mol Biol. 2003 Feb 14;326(2):493–502. pmid:12559917
  49. 49. Yasuda J, Nakao M, Kawaoka Y, Shida H. Nedd4 regulates egress of Ebola virus-like particles from host cells. J Virol. 2003 Sep;77(18):9987–9992. pmid:12941909
  50. 50. Urata S, Noda T, Kawaoka Y, Morikawa S, Yokosawa H, Yasuda J. Interaction of Tsg101 with Marburg Virus VP40 Depends on the PPPY Motif, but Not the PT/SAP Motif as in the Case of Ebola Virus, and Tsg101 Plays a Critical Role in the Budding of Marburg Virus-Like Particles Induced by VP40, NP, and GP. J Virol. 2007 May 1;81(9):4895–4899.
  51. 51. Carpp LN, Galler R, Bonaldo MC. Interaction between the yellow fever virus nonstructural protein NS3 and the host protein Alix contributes to the release of infectious particles. Microbes Infect. 2011 Jan;13(1):85–95. pmid:21044891
  52. 52. Chiou CT, Hu CCA, Chen PH, Liao CL, Lin YL, Wang JJ. Association of Japanese encephalitis virus NS3 protein with microtubules and tumour susceptibility gene 101 (TSG101) protein. J Gen Virol. 2003 Oct;84(Pt 10):2795–2805. pmid:13679614
  53. 53. Tabata K, Arimoto M, Arakawa M, Nara A, Saito K, Omori H, et al. Unique Requirement for ESCRT Factors in Flavivirus Particle Formation on the Endoplasmic Reticulum. Cell Rep. 2016 30;16(9):2339–2347. pmid:27545892
  54. 54. Thepparit C, Khongwichit S, Ketsuwan K, Libsittikul S, Auewarakul P, Smith DR. Dengue virus requires apoptosis linked gene-2-interacting protein X (ALIX) for viral propagation. Virus Res. 2019 Feb;261:65–71. pmid:30599162
  55. 55. Ariumi Y, Kuroki M, Maki M, Ikeda M, Dansako H, Wakita T, et al. The ESCRT system is required for hepatitis C virus production. PLoS ONE. 2011 Jan 11;6(1):e14517. pmid:21264300
  56. 56. Barouch-Bentov R, Neveu G, Xiao F, Beer M, Bekerman E, Schor S, et al. Hepatitis C Virus Proteins Interact with the Endosomal Sorting Complex Required for Transport (ESCRT) Machinery via Ubiquitination To Facilitate Viral Envelopment. MBio. 2016 01;7(6). pmid:27803188
  57. 57. Tamai K, Shiina M, Tanaka N, Nakano T, Yamamoto A, Kondo Y, et al. Regulation of hepatitis C virus secretion by the Hrs-dependent exosomal pathway. Virology. 2012 Jan 20;422(2):377–385. pmid:22138215
  58. 58. Corless L, Crump CM, Griffin SDC, Harris M. Vps4 and the ESCRT-III complex are required for the release of infectious hepatitis C virus particles. J Gen Virol. 2010 Feb;91(Pt 2):362–372. pmid:19828764
  59. 59. Liu CC, Liu YY, Cheng Y, Zhang YN, Zhang J, Liang XD, et al. The ESCRT-I Subunit Tsg101 Plays Novel Dual Roles in Entry and Replication of Classical Swine Fever Virus. J Virol. 2021 Feb 24;95(6):e01928–e01920. pmid:33328308
  60. 60. Liu CC, Liu YY, Zhou JF, Chen X, Chen H, Hu JH, et al. Cellular ESCRT components are recruited to regulate the endocytic trafficking and RNA replication compartment assembly during classical swine fever virus infection. PLoS Pathog. 2022 Feb 4;18(2):e1010294. pmid:35120190
  61. 61. Tran PTH, Chiramel AI, Johansson M, Melik W. Roles of ESCRT Proteins ALIX and CHMP4A and Their Interplay with Interferon-Stimulated Gene 15 during Tick-Borne Flavivirus Infection. J Virol. 2022 Feb 9;96(3):e0162421. pmid:34851141
  62. 62. Feng Z, Hensley L, McKnight KL, Hu F, Madden V, Ping L, et al. A pathogenic picornavirus acquires an envelope by hijacking cellular membranes. Nature. 2013 Apr 18;496(7445):367–371. pmid:23542590
  63. 63. McKnight KL, Xie L, González-López O, Rivera-Serrano EE, Chen X, Lemon SM. Protein composition of the hepatitis A virus quasi-envelope. Proc Natl Acad Sci. 2017 Jun 20;114(25):6587–6592. pmid:28490497
  64. 64. Shirasaki T, Feng H, Duyvesteyn HME, Fusco WG, McKnight KL, Xie L, et al. Nonlytic cellular release of hepatitis A virus requires dual capsid recruitment of the ESCRT-associated Bro1 domain proteins HD-PTP and ALIX. PLoS Pathog. 2022 Aug 15;18(8):e1010543.
  65. 65. González-López O, Rivera-Serrano EE, Hu F, Hensley L, McKnight KL, Ren J, et al. Redundant Late Domain Functions of Tandem VP2 YPX3L Motifs in Nonlytic Cellular Egress of Quasi-enveloped Hepatitis A Virus. J Virol. 2018 Nov 12;92(23):e01308–e01318. pmid:30232181
  66. 66. Diaz A, Zhang J, Ollwerther A, Wang X, Ahlquist P. Host ESCRT proteins are required for bromovirus RNA replication compartment assembly and function. PLoS Pathog. 2015 Mar;11(3):e1004742. pmid:25748299
  67. 67. Jiang W, Ma P, Deng L, Liu Z, Wang X, Liu X, et al. Hepatitis A virus structural protein pX interacts with ALIX and promotes the secretion of virions and foreign proteins through exosome-like vesicles. J Extracell Vesicles. 2020. pmid:32082513
  68. 68. Torii S, Orba Y, Sasaki M, Tabata K, Wada Y, Carr M, et al. Host ESCRT factors are recruited during chikungunya virus infection and are required for the intracellular viral replication cycle. J Biol Chem. 2020 Jun 5;295(23):7941–7957. pmid:32341071
  69. 69. Arii J, Watanabe M, Maeda F, Tokai-Nishizumi N, Chihara T, Miura M, et al. ESCRT-III mediates budding across the inner nuclear membrane and regulates its integrity. Nat Commun. 2018 Aug 23;9:3379. pmid:30139939
  70. 70. Arii J, Takeshima K, Maruzuru Y, Koyanagi N, Nakayama Y, Kato A, et al. Role of the arginine cluster in the disordered domain of Herpes Simplex Virus 1 UL34 for the recruitment of ESCRT-III for viral primary envelopment. J Virol [Internet]. 2021 Nov 3 [cited 2022 Jan 12]; Available from: https://journals.asm.org/doi/abs/10.1128/JVI.01704-21 pmid:34730397
  71. 71. Calistri A, Sette P, Salata C, Cancellotti E, Forghieri C, Comin A, et al. Intracellular trafficking and maturation of herpes simplex virus type 1 gB and virus egress require functional biogenesis of multivesicular bodies. J Virol. 2007 Oct;81(20):11468–11478. pmid:17686835
  72. 72. Crump CM, Yates C, Minson T. Herpes simplex virus type 1 cytoplasmic envelopment requires functional Vps4. J Virol. 2007 Jul;81(14):7380–7387. pmid:17507493
  73. 73. Kharkwal H, Smith CG, Wilson DW. Herpes Simplex Virus Capsid Localization to ESCRT-VPS4 Complexes in the Presence and Absence of the Large Tegument Protein UL36p. J Virol. 2016 Aug 15;90(16):7257–7267. pmid:27252536
  74. 74. Calistri A, Munegato D, Toffoletto M, Celestino M, Franchin E, Comin A, et al. Functional Interaction Between the ESCRT-I Component TSG101 and the HSV-1 Tegument Ubiquitin Specific Protease. J Cell Physiol. 2015;230(8):1794–1806. pmid:25510868
  75. 75. Pawliczek T, Crump CM. Herpes simplex virus type 1 production requires a functional ESCRT-III complex but is independent of TSG101 and ALIX expression. J Virol. 2009 Nov;83(21):11254–11264.
  76. 76. Lee CP, Liu PT, Kung HN, Su MT, Chua HH, Chang YH, et al. The ESCRT machinery is recruited by the viral BFRF1 protein to the nucleus-associated membrane for the maturation of Epstein-Barr Virus. PLoS Pathog. 2012 Sep;8(9):e1002904. pmid:22969426
  77. 77. Lee CP, Liu GT, Kung HN, Liu PT, Liao YT, Chow LP, et al. The Ubiquitin Ligase Itch and Ubiquitination Regulate BFRF1-Mediated Nuclear Envelope Modification for Epstein-Barr Virus Maturation. J Virol [Internet]. 2016 Jul 27 [cited 2022 Feb 4]; Available from: https://journals.asm.org/doi/abs/10.1128/JVI.01235-16 pmid:27466427
  78. 78. Chua HH, Lee HH, Chang SS, Lu CC, Yeh TH, Hsu TY, et al. Role of the TSG101 gene in Epstein-Barr virus late gene transcription. J Virol. 2007 Mar;81(5):2459–2471. pmid:17182691
  79. 79. Tandon R, AuCoin DP, Mocarski ES. Human Cytomegalovirus Exploits ESCRT Machinery in the Process of Virion Maturation. J Virol. 2009 Oct;83(20):10797–10807. pmid:19640981
  80. 80. Honeychurch KM, Yang G, Jordan R, Hruby DE. The vaccinia virus F13L YPPL motif is required for efficient release of extracellular enveloped virus. J Virol. 2007 Jul;81(13):7310–7315. pmid:17475658
  81. 81. Huttunen M, Samolej J, Evans RJ, Yakimovich A, White IJ, Kriston-Vizi J, et al. Vaccinia virus hijacks ESCRT-mediated multivesicular body formation for virus egress. Life Sci Alliance. 2021 Aug;4(8):e202000910. pmid:34145027
  82. 82. Lambert C, Döring T, Prange R. Hepatitis B virus maturation is sensitive to functional inhibition of ESCRT-III, Vps4, and gamma 2-adaptin. J Virol. 2007 Sep;81(17):9050–9060. pmid:17553870
  83. 83. Chou SF, Tsai ML, Huang JY, Chang YS, Shih C. The Dual Role of an ESCRT-0 Component HGS in HBV Transcription and Naked Capsid Secretion. PLoS Pathog. 2015 Oct;11(10):e1005123. pmid:26431433
  84. 84. Kian Chua P, Lin MH, Shih C. Potent inhibition of human Hepatitis B virus replication by a host factor Vps4. Virology. 2006 Oct 10;354(1):1–6. pmid:16920176
  85. 85. Watanabe T, Sorensen EM, Naito A, Schott M, Kim S, Ahlquist P. Involvement of host cellular multivesicular body functions in hepatitis B virus budding. Proc Natl Acad Sci U S A. 2007 Jun 12;104(24):10205–10210. pmid:17551004
  86. 86. Read CB, Lind MCH, Chiarelli TJ, Izac JR, Adcox HE. Marconi RT, et al, The Obligate Intracellular Bacterial Pathogen Anaplasma phagocytophilum Exploits Host Cell Multivesicular Body Biogenesis for Proliferation and Dissemination. MBio. 2022 Nov;21:e0296122.
  87. 87. Spera JM, Guaimas F, Czibener C, Ugalde JE. Brucella Egresses from Host Cells Exploiting Multivesicular Bodies. MBio. 2023 Feb 28;14(1):e0333822. pmid:36622142
  88. 88. Miao Y, Li G, Zhang X, Xu H, Abraham SN. A TRP Channel Senses Lysosome Neutralization by Pathogens to Trigger Their Expulsion. Cell. 2015 Jun 4;161(6):1306–1319. pmid:26027738
  89. 89. Mehra A, Zahra A, Thompson V, Sirisaengtaksin N, Wells A, Porto M, et al. Mycobacterium tuberculosis type VII secreted effector EsxH targets host ESCRT to impair trafficking. PLoS Pathog. 2013 Oct;9(10):e1003734.
  90. 90. Mittal E, Skowyra ML, Uwase G, Tinaztepe E, Mehra A. Köster S, et al, Mycobacterium tuberculosis Type VII Secretion System Effectors Differentially Impact the ESCRT Endomembrane Damage Response. MBio. 2018;27:9(6).
  91. 91. Göser V, Kehl A, Röder J, Hensel M. Role of the ESCRT-III complex in controlling integrity of the Salmonella-containing vacuole. Cell Microbiol. 2020 Jun;22(6):e13176. pmid:32017351
  92. 92. Kehl A, Göser V, Reuter T, Liss V, Franke M, John C, et al. A trafficome-wide RNAi screen reveals deployment of early and late secretory host proteins and the entire late endo−/lysosomal vesicle fusion machinery by intracellular Salmonella. PLoS Pathog. 2020 Jul;16(7):e1008220. pmid:32658937
  93. 93. Dukes JD, Lee H, Hagen R, Reaves BJ, Layton AN, Galyov EE, et al. The secreted Salmonella dublin phosphoinositide phosphatase, SopB, localizes to PtdIns(3)P-containing endosomes and perturbs normal endosome to lysosome trafficking. Biochem J. 2006 Apr 15;395(2):239–247. pmid:16396630
  94. 94. Radulovic M, Schink KO, Wenzel EM, Nähse V, Bongiovanni A, Lafont F, et al. ESCRT-mediated lysosome repair precedes lysophagy and promotes cell survival. EMBO J. 2018 Nov 2;37(21):e99753. pmid:30314966
  95. 95. Rivera-Cuevas Y, Mayoral J, Di Cristina M, Lawrence ALE, Olafsson EB, Patel RK, et al. Toxoplasma gondii exploits the host ESCRT machinery for parasite uptake of host cytosolic proteins. PLoS Pathog. 2021 Dec 13;17(12):e1010138. pmid:34898650
  96. 96. Mayoral J, Guevara RB, Rivera-Cuevas Y, Tu V, Tomita T, Romano JD, et al. Dense Granule Protein GRA64 Interacts with Host Cell ESCRT Proteins during Toxoplasma gondii Infection. MBio. 2022 Aug 30;13(4):e0144222. pmid:35730903
  97. 97. Romano JD, Mayoral J, Guevara RB, Rivera-Cuevas Y, Carruthers VB, Weiss LM, et al. Toxoplasma gondii scavenges mammalian host organelles through the usurpation of host ESCRT-III and Vps4A. J Cell Sci. 2023 Feb 15;136(4):jcs260159. pmid:36718630
  98. 98. Guerin A, Corrales RM, Parker ML, Lamarque MH, Jacot D, Hajj HE, et al. Efficient invasion by Toxoplasma depends on the subversion of host protein networks. Nat Microbiol. 2017 Oct;2(10):1358–1366. pmid:28848228
  99. 99. Obita T, Saksena S, Ghazi-Tabatabai S, Gill DJ, Perisic O, Emr SD, et al. Structural basis for selective recognition of ESCRT-III by the AAA ATPase Vps4. Nature. 2007 Oct;449(7163):735–739. pmid:17928861
  100. 100. Hobel CFV, Albers SV, Driessen AJM, Lupas AN. The Sulfolobus solfataricus AAA protein Sso0909, a homologue of the eukaryotic ESCRT Vps4 ATPase. Biochem Soc Trans. 2008 Jan 22;36(1):94–98.
  101. 101. Quemin ERJ, Chlanda P, Sachse M, Forterre P, Prangishvili D, Krupovic M. Eukaryotic-Like Virus Budding in Archaea. mBio [Internet]. 2016 Sep 13 [cited 2021 May 15];7(5). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5021807/ pmid:27624130
  102. 102. Lu Z, Fu T, Li T, Liu Y, Zhang S, Li J, et al. Coevolution of Eukaryote-like Vps4 and ESCRT-III Subunits in the Asgard Archaea. mBio [Internet]. 2020 May 19 [cited 2022 Jan 22]; Available from: https://journals.asm.org/doi/abs/ pmid:32430468
  103. 103. Zaremba-Niedzwiedzka K, Caceres EF, Saw JH, Bäckström D, Juzokaite L, Vancaester E, et al. Asgard archaea illuminate the origin of eukaryotic cellular complexity. Nature. 2017 Jan 19;541(7637):353–358. pmid:28077874
  104. 104. Gao C, Zhuang X, Shen J, Jiang L. Plant ESCRT Complexes: Moving Beyond Endosomal Sorting. Trends Plant Sci. 2017 Nov 1;22(11):986–998. pmid:28867368
  105. 105. Martelli GP, Gallitelli D, Russo M. Tombusviruses. In: Koenig R, editor. The Plant Viruses: Polyhedral Virions with Monopartite RNA Genomes [Internet]. Boston, MA: Springer US; 1988 [cited 2022 Jan 24]. p. 13–72. (The Viruses). Available from: https://doi.org/10.1007/978-1-4613-0921-5_2
  106. 106. McCartney AW, Greenwood JS, Fabian MR, White KA, Mullen RT. Localization of the Tomato Bushy Stunt Virus Replication Protein p33 Reveals a Peroxisome-to-Endoplasmic Reticulum Sorting Pathway[W]. Plant Cell. 2005 Dec 1;17(12):3513–3531.
  107. 107. Nagy PD, Strating JRPM, van Kuppeveld FJM. Building Viral Replication Organelles: Close Encounters of the Membrane Types. Condit RC, editor. PLoS Pathog. 2016 Oct 27;12(10):e1005912. pmid:27788266
  108. 108. Jin X, Cao X, Wang X, Jiang J, Wan J, Laliberté JF, et al. Three-Dimensional Architecture and Biogenesis of Membrane Structures Associated with Plant Virus Replication. Front Plant Sci. 2018 Jan 30;9:57. pmid:29441085
  109. 109. Nagy PD. Yeast as a Model Host to Explore Plant Virus-Host Interactions. Annu Rev Phytopathol. 2008 Sep 1;46(1):217–242.
  110. 110. Nagy PD, Pogany J, Lin JY. How yeast can be used as a genetic platform to explore virus–host interactions: from ‘omics’ to functional studies. Trends Microbiol. 2014 Jun 1;22(6):309–316. pmid:24647076
  111. 111. Panavas T, Serviene E, Brasher J, Nagy PD. Yeast genome-wide screen reveals dissimilar sets of host genes affecting replication of RNA viruses. Proc Natl Acad Sci U S A. 2005 May 17;102(20):7326–7331. pmid:15883361
  112. 112. Barajas D, Nagy PD. Ubiquitination of tombusvirus p33 replication protein plays a role in virus replication and binding to the host Vps23p ESCRT protein. Virology. 2010 Feb 20;397(2):358–368. pmid:20004458
  113. 113. Quinney KB, Frankel EB, Shankar R, Kasberg W, Luong P, Audhya A. Growth factor stimulation promotes multivesicular endosome biogenesis by prolonging recruitment of the late-acting ESCRT machinery. Proc Natl Acad Sci. 2019 Apr 2;116(14):6858–6867. pmid:30894482
  114. 114. Wang X, Ahlquist P. Brome Mosaic Virus. Encycl Virol. 2008;381.
  115. 115. Cui Y, Gao J, He Y, Jiang L. Plant extracellular vesicles. Protoplasma. 2020 Jan;257(1):3–12. pmid:31468195
  116. 116. Hansen LL, Nielsen ME. Plant exosomes: using an unconventional exit to prevent pathogen entry? J Exp Bot. 2018 Jan 1;69(1):59–68.
  117. 117. Cai Q, Qiao L, Wang M, He B, Lin FM, Palmquist J, et al. Plants send small RNAs in extracellular vesicles to fungal pathogen to silence virulence genes. Science. 2018 Jun 8;360(6393):1126–1129. pmid:29773668
  118. 118. Rutter BD, Innes RW. Extracellular Vesicles Isolated from the Leaf Apoplast Carry Stress-Response Proteins. Plant Physiol. 2017 Jan;173(1):728–741. pmid:27837092
  119. 119. Votteler J, Sundquist WI. Virus Budding and the ESCRT Pathway. Cell Host Microbe [Internet]. 2013 Sep 11 [cited 2021 May 9];14(3). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3819203/ pmid:24034610
  120. 120. Demirov DG, Freed EO. Retrovirus budding. Virus Res. 2004 Dec 1;106(2):87–102. pmid:15567490
  121. 121. Welker L, Paillart JC, Bernacchi S. Importance of Viral Late Domains in Budding and Release of Enveloped RNA Viruses. Viruses. 2021 Aug 6;13(8):1559. pmid:34452424
  122. 122. Meng B, Lever AML. The Interplay between ESCRT and Viral Factors in the Enveloped Virus Life Cycle. Viruses [Internet]. 2021 Feb 20 [cited 2021 Mar 8];13(2). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7923801/ pmid:33672541
  123. 123. McDonald B, Martin-Serrano J. No strings attached: the ESCRT machinery in viral budding and cytokinesis. J Cell Sci. 2009 Jul 1;122(13):2167–2177. pmid:19535732
  124. 124. Bell NM, Lever AML. HIV Gag polyprotein: processing and early viral particle assembly. Trends Microbiol. 2013 Mar 1;21(3):136–144. pmid:23266279
  125. 125. Cashikar AG, Shim S, Roth R, Maldazys MR, Heuser JE, Hanson PI. Structure of cellular ESCRT-III spirals and their relationship to HIV budding. Sundquist W, editor. elife. 2014 May 30;3:e02184. pmid:24878737
  126. 126. Prescher J, Baumgärtel V, Ivanchenko S, Torrano AA, Bräuchle C, Müller B, et al. Super-resolution imaging of ESCRT-proteins at HIV-1 assembly sites. PLoS Pathog. 2015 Feb;11(2):e1004677. pmid:25710462
  127. 127. Jouvenet N, Zhadina M, Bieniasz PD, Simon SM. Dynamics of ESCRT protein recruitment during retroviral assembly. Nat Cell Biol. 2011 Apr;13(4):394–401. pmid:21394083
  128. 128. Bleck M, Itano MS, Johnson DS, Thomas VK, North AJ, Bieniasz PD, et al. Temporal and spatial organization of ESCRT protein recruitment during HIV-1 budding. Proc Natl Acad Sci U S A. 2014 Aug 19;111(33):12211–12216. pmid:25099357
  129. 129. Gupta S, Bromley J, Saffarian S. High-speed imaging of ESCRT recruitment and dynamics during HIV virus like particle budding. PLoS ONE. 2020 Sep 4;15(9):e0237268. pmid:32886660
  130. 130. Baumgärtel V, Ivanchenko S, Dupont A, Sergeev M, Wiseman PW, Kräusslich HG, et al. Live-cell visualization of dynamics of HIV budding site interactions with an ESCRT component. Nat Cell Biol. 2011 Apr;13(4):469–474. pmid:21394086
  131. 131. den Boon JA, Diaz A, Ahlquist P. Cytoplasmic Viral Replication Complexes. Cell Host Microbe. 2010 Jul 22;8(1):77–85. pmid:20638644
  132. 132. Mackenzie J. Wrapping Things up about Virus RNA Replication. Traffic Cph Den. 2005 Nov;6(11):967–977. pmid:16190978
  133. 133. Arakawa M, Morita E. Flavivirus Replication Organelle Biogenesis in the Endoplasmic Reticulum: Comparison with Other Single-Stranded Positive-Sense RNA Viruses. Int J Mol Sci. 2019 May 11;20(9):E2336. pmid:31083507
  134. 134. Paul D, Bartenschlager R. Flaviviridae Replication Organelles: Oh, What a Tangled Web We Weave. Annu Rev Virol. 2015 Nov;2(1):289–310. pmid:26958917
  135. 135. Bigalke JM, Heldwein EE. Nuclear Exodus: Herpesviruses Lead the Way. Annu Rev Virol. 2016 Sep 29;3(1):387–409. pmid:27482898
  136. 136. Mettenleiter TC, Klupp BG, Granzow H. Herpesvirus assembly: a tale of two membranes. Curr Opin Microbiol. 2006 Aug 1;9(4):423–429. pmid:16814597
  137. 137. Turcotte S, Letellier J, Lippé R. Herpes Simplex Virus Type 1 Capsids Transit by the trans-Golgi Network, Where Viral Glycoproteins Accumulate Independently of Capsid Egress. J Virol [Internet]. 2005 Jul [cited 2022 Feb 10]; Available from: https://journals.asm.org/doi/abs/10.1128/JVI.79.14.8847-8860.2005 pmid:15994778
  138. 138. Butt BG, Owen DJ, Jeffries CM, Ivanova L, Hill CH, Houghton JW, et al. Insights into herpesvirus assembly from the structure of the pUL7:pUL51 complex. elife. 2020 May 11;9:e53789. pmid:32391791
  139. 139. Truchan HK, Cockburn CL, Hebert KS, Magunda F, Noh SM, Carlyon JA. The Pathogen-Occupied Vacuoles of Anaplasma phagocytophilum and Anaplasma marginale Interact with the Endoplasmic Reticulum. Front Cell Infect Microbiol. 2016 Mar 1;6:22.
  140. 140. Sachdeva K, Sundaramurthy V. The Interplay of Host Lysosomes and Intracellular Pathogens. Front Cell Infect Microbiol. 2020 Nov 20;10:595502. pmid:33330138
  141. 141. Heinzen RA, Scidmore MA, Rockey DD, Hackstadt T. Differential interaction with endocytic and exocytic pathways distinguish parasitophorous vacuoles of Coxiella burnetii and Chlamydia trachomatis. Infect Immun. 1996 Mar;64(3):796–809.
  142. 142. Philips JA, Porto MC, Wang H, Rubin EJ, Perrimon N. ESCRT factors restrict mycobacterial growth. Proc Natl Acad Sci U S A. 2008 Feb 26;105(8):3070–3075. pmid:18287038
  143. 143. Griffith MB, Pearce CS, Heaslip AT. Dense granule biogenesis, secretion, and function in Toxoplasma gondii. J Eukaryot Microbiol. 2022 Nov;69(6):e12904. pmid:35302693
  144. 144. Carruthers VB, Sibley LD. Sequential protein secretion from three distinct organelles of Toxoplasma gondii accompanies invasion of human fibroblasts. Eur J Cell Biol. 1997 Jun;73(2):114–123. pmid:9208224
  145. 145. Wang Y, Sangaré LO, Paredes-Santos TC, Saeij JPJ. Toxoplasma Mechanisms for Delivery of Proteins and Uptake of Nutrients Across the Host-Pathogen Interface. Annu Rev Microbiol. 2020 Sep 8;74:567–586. pmid:32680452
  146. 146. Dou Z, McGovern OL, Di Cristina M, Carruthers VB. Toxoplasma gondii ingests and digests host cytosolic proteins. mBio. 2014 Jul 15;5(4):e01188–e01114. pmid:25028423
  147. 147. Romano JD, Nolan SJ, Porter C, Ehrenman K, Hartman EJ, R Ching H, et al. The parasite Toxoplasma sequesters diverse Rab host vesicles within an intravacuolar network. J Cell Biol. 2017 Oct 25;216(12):4235–4254. pmid:29070609
  148. 148. Cygan AM, Jean Beltran PM, Mendoza AG, Branon TC, Ting AY, Carr SA, et al. Proximity-Labeling Reveals Novel Host and Parasite Proteins at the Toxoplasma Parasitophorous Vacuole Membrane. MBio. 2021 Dec 21;12(6):e0026021. pmid:34749525
  149. 149. Ritter AT, Shtengel G, Xu CS, Weigel A, Hoffman DP, Freeman M, et al. ESCRT-mediated membrane repair protects tumor-derived cells against T cell attack. Science. 2022 Apr 22;376(6591):377–382. pmid:35446649
  150. 150. Yang Y, Wang M, Zhang YY, Zhao SZ, Gu S. The endosomal sorting complex required for transport repairs the membrane to delay cell death. Front Oncol. 2022 Oct 18;12:1007446. pmid:36330465
  151. 151. Rühl S, Shkarina K, Demarco B, Heilig R, Santos JC, Broz P. ESCRT-dependent membrane repair negatively regulates pyroptosis downstream of GSDMD activation. Science. 2018 Nov 23;362(6417):956–960. pmid:30467171
  152. 152. Westman J, Plumb J, Licht A, Yang M, Allert S, Naglik JR, et al. Calcium-dependent ESCRT recruitment and lysosome exocytosis maintain epithelial integrity during Candida albicans invasion. Cell Rep. 2022 Jan 4;38(1):110187.
  153. 153. Wegner CS, Rodahl LMW, Stenmark H. ESCRT Proteins and Cell Signalling. Traffic. 2011;12(10):1291–1297. pmid:21518165
  154. 154. Kuchitsu Y, Mukai K, Uematsu R, Takaada Y, Shinojima A, Shindo R, et al. STING signalling is terminated through ESCRT-dependent microautophagy of vesicles originating from recycling endosomes. Nat Cell Biol. 2023 Mar;25(3):453–466. pmid:36918692
  155. 155. Vardhana S, Choudhuri K, Varma R, Dustin ML. Essential role of Ubiquitin and TSG101 in formation and function of the central supramolecular activation cluster. Immunity. 2010 Apr 23;32(4):531–540.
  156. 156. Choudhuri K, Llodrá J, Roth EW, Tsai J, Gordo S, Wucherpfennig KW, et al. Polarized release of T-cell-receptor-enriched microvesicles at the immunological synapse. Nature. 2014 Mar 6;507(7490):118–123. pmid:24487619
  157. 157. Kvalvaag A, Valvo S, Céspedes PF, Saliba DG, Kurz E, Korobchevskaya K, et al. Clathrin mediates both internalization and vesicular release of triggered T cell receptor at the immunological synapse. Proc Natl Acad Sci U S A. 2023 Feb 7;120(6):e2211368120. pmid:36730202
  158. 158. Gros M, Segura E, Rookhuizen DC, Baudon B, Heurtebise-Chrétien S, Burgdorf N, et al. Endocytic membrane repair by ESCRT-III controls antigen export to the cytosol during antigen cross-presentation. Cell Rep. 2022 Aug 16;40(7):111205. pmid:35977488
  159. 159. Croce C, Garrido F, Dinamarca S, Santi-Rocca J, Marion S, Blanchard N, et al. Efficient Cholesterol Transport in Dendritic Cells Defines Optimal Exogenous Antigen Presentation and Toxoplasma gondii Proliferation. Front Cell Dev Biol [Internet]. 2022 [cited 2022 Nov 8];10. Available from: https://www.frontiersin.org/articles/10.3389/fcell.2022.837574 pmid:35309938
  160. 160. Bohannon KP, Hanson PI. ESCRT puts its thumb on the nanoscale: Fixing tiny holes in endolysosomes. Curr Opin Cell Biol. 2020 Aug;65:122–130. pmid:32731154
  161. 161. Sergeeva OA, van der Goot FG. Kicking Out Pathogens in Exosomes. Cell. 2015 Jun 4;161(6):1241–1242. pmid:26046431
  162. 162. Leung KF, Dacks JB, Field MC. Evolution of the multivesicular body ESCRT machinery; retention across the eukaryotic lineage. Traffic Cph Den. 2008 Sep;9(10):1698–1716.
  163. 163. Cruz Camacho A, Alfandari D, Kozela E, Regev-Rudzki N. Biogenesis of extracellular vesicles in protozoan parasites: The ESCRT complex in the trafficking fast lane? PLoS Pathog. 2023 Feb;19(2):e1011140. pmid:36821560
  164. 164. Avalos-Padilla Y, Georgiev VN, Lantero E, Pujals S, Verhoef R, N Borgheti-Cardoso L, et al. The ESCRT-III machinery participates in the production of extracellular vesicles and protein export during Plasmodium falciparum infection. PLoS Pathog. 2021 Apr;17(4):e1009455. pmid:33798247
  165. 165. Nandakumar R, Tschismarov R, Meissner F, Prabakaran T, Krissanaprasit A, Farahani E, et al. Intracellular bacteria engage a STING-TBK1-MVB12b pathway to enable paracrine cGAS-STING signalling. Nat Microbiol. 2019 Apr;4(4):701–713. pmid:30804548
  166. 166. Bache KG, Brech A, Mehlum A, Stenmark H. Hrs regulates multivesicular body formation via ESCRT recruitment to endosomes. J Cell Biol. 2003 Aug 4;162(3):435–442. pmid:12900395