Next Article in Journal
Outcomes Using Focused Shockwave for Treatment of Bone Stress Injury in Runners
Previous Article in Journal
Prediction of COVID-19 Using a WOA-BILSTM Model
Previous Article in Special Issue
Cancer Drug Delivery Systems Using Bacterial Toxin Translocation Mechanisms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gaining New Insights into Fundamental Biological Pathways by Bacterial Toxin-Based Genetic Screens

1
State Key Laboratory of Natural and Biomimetic Drugs, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100191, China
2
Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA
3
Department of Microbiology and Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
*
Author to whom correspondence should be addressed.
Bioengineering 2023, 10(8), 884; https://doi.org/10.3390/bioengineering10080884
Submission received: 15 May 2023 / Revised: 11 July 2023 / Accepted: 22 July 2023 / Published: 25 July 2023
(This article belongs to the Special Issue Advances in Bacterial Toxin-Based Platforms)

Abstract

:
Genetic screen technology has been applied to study the mechanism of action of bacterial toxins—a special class of virulence factors that contribute to the pathogenesis caused by bacterial infections. These screens aim to identify host factors that directly or indirectly facilitate toxin intoxication. Additionally, specific properties of certain toxins, such as membrane interaction, retrograde trafficking, and carbohydrate binding, provide robust probes to comprehensively investigate the lipid biosynthesis, membrane vesicle transport, and glycosylation pathways, respectively. This review specifically focuses on recent representative toxin-based genetic screens that have identified new players involved in and provided new insights into fundamental biological pathways, such as glycosphingolipid biosynthesis, protein glycosylation, and membrane vesicle trafficking pathways. Functionally characterizing these newly identified factors not only expands our current understanding of toxin biology but also enables a deeper comprehension of fundamental biological questions. Consequently, it stimulates the development of new therapeutic approaches targeting both bacterial infectious diseases and genetic disorders with defects in these factors and pathways.

1. Introduction

Bacterial exotoxins are special virulence factors responsible for many infectious diseases caused by bacterial pathogens [1,2]. Once being produced and released, these toxins autonomously target host cells by recognizing cell surface receptors through their highly specific receptor-binding moieties. Some toxins, such as membrane-damaging toxins and pore-forming toxins, act directly on the plasma membrane. In contrast, the more dangerous toxins, including retrograde trafficking toxins and single-chain toxins, deliver their enzymatic moieties across the membrane to act on their intracellular substrates (Figure 1). Extensive discussions have focused on the identification, mechanisms, and significance of these toxins [3,4,5,6,7,8,9].
Throughout the intoxication process, toxins exploit multiple host machinery to facilitate their attachment, uptake, translocation, and activation (Figure 1). Identifying and characterizing these host factors and pathways can provide important comprehension of the basic aspects of host–toxin interactions and aid in discovering novel anti-toxin therapeutics. Moreover, some newly identified host factors (excluding receptors) do not physically interact with toxins. Instead, they may be involved in complex cellular processes that are indirectly utilized by the toxins. In this case, toxins can serve as valuable probes to elucidate the roles of these novel players in fundamental biological pathways.
A key task in genetic analysis is to connect a specific phenotype with a gene. Reverse genetic approaches firstly generate a precise genetic perturbation and then track the consequential phenotypes. In contrast, forward genetic screens firstly modulate a panel of genes and then identify the gene responsible for a phenotype of interest [10]. These screens have discovered fundamental biological pathways in model organisms such as yeast, worms, fish, rodents, and humans [11,12].
The comprehensive genome-wide forward genetic screen is a straightforward and unbiased strategy to uncover unknown host factors for a toxin. This is because the surviving cells after toxin treatment may carry mutations in specific genes that have been involved in the toxin-induced killing (Figure 2). Historically, gain-of-function screens were introduced to the toxin field earlier than loss-of-function approaches because the classic technologies, such as complementary DNA (cDNA) expression cloning [13,14], could efficiently lead to ectopic overexpression of genes in mammalian cells in a high-throughput manner. The cellular receptors of many important toxins, such as diphtheria toxin (DT), were identified using this strategy [15]. Recently, technological advancements such as RNA interference (RNAi) [16,17], retrovirus-based insertional mutagenesis in haploid cells [18,19], and the recently developed CRISPR (clustered regularly interspaced short palindromic repeat) system [20,21] have made loss-of-function screens more popular. In particular, the CRISPR system, consisting of CRISPR-associated Cas9 nuclease and single guide RNA (sgRNA), has revolutionized the field of genetic screen due to its ability to induce strong loss-of-function mutations (knockout) at precise loci on both alleles in diploid genomes [22,23]. As an alternative approach, the CRISPR repression (CRISPRi) system blocks the transcription and achieves mild loss-of-function (knockdown) [24,25]. On the other hand, the emerging gain-of-function screen relies on CRISPR activation (CRISPRa) technology [26], which is also valuable for studying host–toxin interactions (Figure 2).
The RNAi screen is easy to handle and can be applied to multiple cell lines to achieve high genome coverage. However, this post-transcriptional gene repression approach has significant off-target effects and may lead to hypomorphic mutations, where the targeted genes are partially suppressed [27]. Compared to RNAi, CRISPRi affects chromatin and results in more effective knockdown outcomes [26]. The retrovirus-based insertional screen offers a highly efficient method for generating genome-wide loss-of-function. However, it requires haploid or nearly haploid cells, and the insertion sites have a virus-dependent preference [28], which may result in incomplete genome coverage. In gain-of-function approaches, cDNA expression cloning is widely used, but the expression levels of each gene are not well controlled, leading to inconsistencies in phenotypic outcomes [14]. In contrast, the CRISPRa approach offers a more controlled method to specifically activate gene expression [29]. It should be noted that all the Cas9/sgRNA-based mutagenesis strategies may have a certain level of off-target effects [30].
The genome-wide CRISPR-Cas9-mediated loss-of-function screen is the most widely used strategy with multiple advantages. Notably, although complete knockout mutations can be generated at most alleles, some cells may retain a copy of the allele with partial function generated by non-frameshift mutations [22]. A typical screen begins through generating a knockout cell library by introducing a sgRNA library (e.g., the GeCKO-v2 library [20]) into the Cas9-expressing cells. Subsequently, the cells are exposed to toxins as a phenotypic selection, usually through multiple rounds, to minimize contaminants. The surviving cells at the end of the selection can be expanded and analyzed using advanced sequencing techniques to determine the specific genes involved (Figure 2) [31,32,33,34,35].
This review specifically focuses on the recent representative genetic screens that relied on classic bacterial toxins (e.g., Shiga toxins, cholera toxin, ricin, diphtheria toxin, Pseudomonas exotoxin A, anthrax toxin, large clostridial toxins, and pore-forming toxins) as screening stresses. These screens identified and characterized host factors that indirectly interact with toxins but play important roles in fundamental biological pathways such as glycosphingolipid biosynthesis, protein glycosylation, membrane vesicle trafficking, and other unique pathways. The objective of this review is to inspire innovative approaches that utilize toxin-based platforms to make fundamental breakthroughs on basic biological questions beyond a deep understanding of the toxin’s mechanism of action.

2. Factors Required for the Biosynthesis of Glycosphingolipids

As integral components of the cell membrane, glycosphingolipids (GSLs) consist of a glycosidically bound carbohydrate moiety and a lipid moiety known as ceramide (Figure 3). The carbohydrate moieties of GSLs can interact with other carbohydrates or proteins, serving as the molecular basis of cell–cell recognition and initiating cellular activities such as immune response, cell proliferation, and apoptosis [36,37]. Additionally, GSL-organized microdomains on the plasma membrane provide a molecular platform for clustering the proteins involved in signal transduction [36,37]. Although GSLs are essential for tissue development, excessive accumulation of GSLs can cause a class of inherited disorders called sphingolipidoses (e.g., Fabry disease, Gaucher disease, and Niemann–Pick disease) [38,39]. Therefore, a deep understanding of the GSL metabolism pathways, encompassing both biosynthesis and degradation, holds promise for developing therapeutic approaches.
The de novo biosynthesis of GSLs (Figure 3) in mammals starts with ceramide, which is synthesized on the endoplasmic reticulum (ER) membrane through four steps of biosynthesis, originally from serine and palmitoyl-CoA. Ceramide is then transported to the Golgi apparatus, where a series of glycosyltransferases catalyze the transfer reactions of carbohydrate moieties between donor and acceptor molecules [41]. UGCG (UDP-glucose ceramide glucosyltransferase) transfers UDP-glucose onto ceramide and generates glucosylceramide (GlcCer) on the cytosolic side of the Golgi. Within the Golgi lumen, B4GALT5 (β-1,4-galactosyltransferase 5) then transfers UDP-galactose onto GlcCer, producing lactosylceramide (LacCer), which is the shared precursor for most of the complex GSLs, such as globosides, lactosides, and gangliosides (Figure 3) [37]. SLC35A2 (solute carrier family 35 member A2) also plays a role in this pathway by transporting UDP-galactose from the cytosol into the Golgi lumen. In addition to de novo synthesis, ceramide can be generated through the salvage pathway (Figure 3) by recycling complex GSLs or re-acylating sphingosine [42,43].
Some bacterial toxins, such as Shiga toxins (Stxs), cholera toxin (Ctx), Escherichia coli heat-labile enterotoxin, tetanus neurotoxin (TeNT), and botulinum neurotoxins (BoNTs), are natural probes for GSLs since they specifically recognize GSLs as cellular receptors [44]. The recognition of GSLs by specific bacterial toxins offers a straightforward approach for conducting toxin-based genetic screens to comprehensively explore the biosynthesis pathway of GSLs. This strategy was challenging to achieve in the past due to limitations associated with traditional techniques. In this context, we focus on the recent screens involving two representative toxins, Stxs and Ctx, which recognize two distinct GSLs: globotriaosylceramide (Gb3, also known as CD77) and monosialotetrahexosylganglioside (GM1) as the receptor, respectively (Figure 3 and Table 1).

2.1. The Biology of Stxs

The Stx family includes the prototype Stx from Shigella dysenteriae and related Shiga-like toxins Stx1 and Stx2, produced by enterohemorrhagic Escherichia coli (EHEC) [45]. Stx1 differs from Stx by only one amino acid residue, whereas Stx2 represents distinct serotypes with ~56% sequence identity compared to Stx [45,46]. Belonging to the AB5 toxin superfamily, the Stx family is composed of an A chain (32 kDa) and five identical B chains (7.7 kDa each). The A chain is the enzymatic domain, which acts as an N-glycosidase that cleaves the host ribosomal RNA, while the five B chains form a pentameric ring and serve as the receptor binding domain. The A chain is connected to the B chain by inserting its C-terminus into the central pore of the B chain pentamer.
Once Stxs bind to the cellular receptor and enter cells through endocytosis by either clathrin-dependent or -independent pathways, they are sorted into the retrograde trafficking route and enter the trans-Golgi network (TGN). The A chain is processed by the host protease furin and cleaved into the enzymatic part A1 (27.5 kDa) and the B chain connecting part A2 (4.5 kDa). The A1 and A2 remain connected through an intramolecular disulfide bond between cysteines 242 and 261 residues. Stxs are further transported into the lumen of the ER, where the disulfide bond is reduced. The A1 part then crosses the ER membrane and enters the cytoplasm, utilizing the host ER-associated protein degradation (ERAD) machinery. The cytosolic Stxs eventually shut down protein synthesis by digesting ribosomal RNA and causing cell death [45,46].
The Stx B-chain pentamer specifically recognizes the carbohydrate moiety of Gb3 as its receptor [45,46,47,48]. The crystal structure suggests that each Stx B chain contains three Gb3 binding sites. Thus, one Stx holotoxin could maximally cluster fifteen Gb3 molecules on the cell surface [48]. Gb3 is the first member of the globo-series GSLs. The synthesis of Gb3 by transferring UDP-galactose onto LacCer is catalyzed by A4GALT (α-1,4-galactosyltransferase, also known as Gb3 synthetase, Figure 3) [49,50]. The expression of Gb3 in humans is highly restricted to the kidney, nervous system, microvascular endothelium, and a subset of germinal center B cells. In contrast, most other cell types do not express detectable levels of Gb3 [51,52,53,54]. The kidney-enriched Gb3 is responsible for the life-threatening post-diarrheal hemolytic uremic syndrome (D+HUS) induced by EHEC infection [46,55]. On the other hand, Gb3 lysosomal accumulation leads to a type of sphingolipidoses known as Fabry disease, which is the consequence of the loss-of-function of a lysosomal enzyme called α-galactosidase A, which is responsible for the degradation of Gb3 [56]. Enzyme replacement therapy is the only reliable treatment for Fabry disease nowadays [57,58]. In contrast, substrate reduction therapy is another promising approach involving the inhibition of Gb3 biosynthesis using small-molecule drugs such as ceramide analogs or imino-sugars, which holds promise as an alternative approach [38].

2.2. Genetic Screens for Stxs

In 2018, Tian et al. reported the first CRISPR-Cas9-mediated genome-wide screen for Stx1 and Stx2 [31]. The screen was conducted using the human bladder carcinoma 5637 cell line in a loss-of-function manner. The majority of the top-ranked hits overlapped between the Stx1 and Stx2 screens. Five top-ranked genes were key factors in the established Gb3 biosynthesis pathway: SPTSSA, UGCG, B4GALT5, A4GALT, and SLC35A2. Particularly, SPTSSA is a component of the serine palmitoyltransferase (SPT) complex on the ER membrane, which catalyzes the rate-limiting step in ceramide generation [37]. Other notable top-ranked hits shared by both screens include UGP2 and SPPL3. UGP2 (UDP-glucose pyrophosphorylase 2) is the key enzyme that produces UDP-glucose, the substrate for GlcCer synthesis. SPPL3 (signal peptide peptidase-like 3) is a Golgi-localized protease implicated in the cleavage and activation of many glycosyltransferases [59,60]. Tian et al. then focused on investigating the other three newly identified factors, LAPTM4A, TMEM165, and TM9SF2 [31].
LAPTM4A (lysosomal-associated protein transmembrane 4 A) was identified as a top hit in the screens (ranking No. 2 in the Stx1 screen and No. 1 in the Stx2 screen). However, its function had not been well characterized. Tian et al. found that knocking out LAPTM4A phenotypically mimicked knocking out A4GALT (Gb3 synthetase) in four aspects: (1) Both dramatically increased the cell resistance to Stx1 and Stx2, but not Ctx; (2) both abolished Stx cell surface binding but had no effect on Ctx binding; (3) both greatly reduced the expression level of Gb3, as measured by the mass spectrometry-based lipidomic assay; and (4) both induced the accumulation of the Gb3 precursor LacCer. However, the Golgi localization and the expression level of A4GALT were not altered in the LAPTM4A-knockout cells. Additionally, these properties of LAPTM4A are not shared with those of its homolog, LAPTM4B [31]. LAPTM4A is a small protein with 233 residues and four transmembrane domains, initially reported as an endosomal/lysosomal protein [61,62]. Tian et al. found that LAPTM4A is predominantly localized in the Golgi in multiple cell lines and physically interacts with A4GALT. Through the investigation of membrane topology and comparison of a panel of LAPTM4A/LAPTM4B chimeric proteins, the second lumenal domain of LAPTM4A was demonstrated to be critical for its function [31]. These results indicate that LAPTM4A is likely involved in the last step of Gb3 biosynthesis by serving as an essential co-factor for A4GALT’s enzymatic activity. However, the molecular basis of the interaction between LAPTM4A and A4GALT remains to be established.
TMEM165 (transmembrane protein 165) encodes a multi-pass transmembrane protein that is Golgi-localized and has been proposed as a transporter for manganese ions (Mn2+) [63]. TMEM165 is critical for maintaining Mn2+ hemostasis, and its mutations have been linked to human disorders with defects in glycosylation [64,65], as Mn2+ is required for many Golgi-localized glycosyltransferases. Tian et al. found that TMEM165-deficient cells were more resistant and had lower cell surface binding to Stx and Ctx. In contrast to LAPTM4A, TMEM165-deficient cells had lower levels of Gb3 and Gb3 precursors and gangliosides, suggesting that TMEM165 affects the biosynthesis of GSLs globally. Consistent with previous reports, Tian et al. confirmed the Golgi localization of TMEM165. Furthermore, the downsides of TMEM165 deficiency could be rescued by supplementing extra Mn2+, and TMEM165-deficient cells showed lower tolerance to Mn2+-induced cytotoxicity [31]. These findings experimentally confirmed the role of TMEM165 in regulating Mn2+ homeostasis.
TM9SF2 (transmembrane 9 superfamily member 2) encodes a highly conserved but poorly characterized multi-pass transmembrane protein that has been reported to have endosomal or Golgi localization [66,67]. It has also been associated with multiple glycosylation pathways, including heparan sulfate proteoglycan biosynthesis [67]. Tian et al. verified the Golgi-localization of TM9SF2 in multiple cell lines and found that TM9SF2-knockout cells had a lower level of surface heparan sulfate. Similar to TMEM165, TM9SF2-knockout cells expressed a lower level of GSLs, indicating that loss of TM9SF2 causes a global disruption in GSL biosynthesis and contributes to the resistance to both Stx and Ctx [31]. The detailed mechanism of how TM9SF2 is involved in glycosylation, whether similar to TMEM165 [54], remains to be established.
In 2019, Yamaji et al. reported an independent CRISPR screen for Stx1 in HeLa cells [68]. The screen once again identified well-established genes involved in GSL biosynthesis and the novel factors LAPTM4A, TMEM165, and TM9SF2. Using radioisotope labeling and thin-layer chromatography, Yamaji et al. demonstrated the requirement for LAPTM4A but not LAPTM4B in the last step of Gb3 synthesis. They further provided direct evidence showing that the enzymatic activity of A4GALT in cell lysates was greatly reduced when LAPTM4A was absent [68]. Yamaji et al. also showed that TM9SF2 is involved in Gb3 biosynthesis (likely through A4GALT) by its conserved C-terminus across all transmembrane 9 superfamily members (TM9SF1, TM9SF2, TM9SF3, and TM9SF4) [68]. In 2021, the same group reported a related screen on Vero cells derived from green monkeys, and the knockout was generated by a library targeting the human genome [69]. Although the degree of gene enrichment was less than the compatible screen in human cells, this screen still identified major players in the GSLs pathway, including LAPTM4A and TM9SF2 [69]. In 2020, Majumder et al. performed a similar screen on HeLa cells [70]. In addition to the same set of factors (including LAPTM4A and TM9SF2), a transcription factor, AHR (aryl hydrocarbon receptor), was uniquely identified, which may regulate Gb3 biosynthesis by regulating the expression of several known factors such as SPTSSA [70].
In 2018, Pacheco et al. conducted a unique screen directly using EHEC. This carries an additional virulence factor besides Stx and is known as the type III secretion system (T3SS) in the human intestinal epithelial HT29 cell line. HT29 cells were chosen because they are sensitive to EHEC co-culture but resistant to purified Stx [71]. Interestingly, many of the identified genes in this screen were involved in Gb3 biosynthesis (including LAPTM4A and TM9SF2), suggesting the potential role of Gb3 in the virulence of both Stx and T3SS. This finding further emphasizes that targeting factors in the GSL biosynthesis pathway is promising for designing new drugs to combat Stx and EHEC infections [71].
Another unique screen was reported by Kono et al., which specially focused on the ceramide salvage pathway [72]. The screen was carried out in HeLa cells with a de novo ceramide synthesis defect by knocking out the key gene SPTLC1 (serine palmitoyltransferase long chain base subunit 1). The SPTLC1-knockout cells are unable to generate 3-Keto-dihydrosphingosine from serine and palmitoyl-CoA. Then, sphingosine-1-phosphate (S1P) was added to the culture medium to activate the salvage pathway and restore the expression of Gb3. The CRISPR screen using Stx2 as killing stress under this condition successfully identified several important genes. These include ceramide synthase CERS2, well-known genes in the Gb3 biosynthesis pathway (UGCG, B4GALT5, A4GALT, and SLC35A2), and three newly identified factors (LAPTM4A, TMEM165, and TM9SF2). Additionally, two phosphatases, PLPP3 (phospholipid phosphatase 3, also known as PPAP2B) and SGPP1 (S1P phosphatase 1), were also identified through this screening process [72]. Kono et al. found that the cell surface-expressed PLPP3 is important for the uptake of extracellular S1P by dephosphorylating S1P into sphingosine. Then, the cellular sphingosine is rephosphorylated to S1P and further dephosphorylated by SGPP1 for ceramide synthesis (Figure 3) [72].

2.3. Ctx and the Related Screens

Ctx is the major virulence factor produced by toxigenic strains of Vibrio cholerae [73]. Similar to Stxs, Ctx also belongs to the AB5 toxin superfamily and shows a similar overall architecture. Upon binding to the cell surface and endocytosis, Ctx undergoes retrograde trafficking and releases its enzymatic A chain across the ER membrane [74]. The cytosol-exposed A chain deactivates the GTP hydrolase activity of the GS alpha subunit through an ADP-ribosylation reaction and causes the continuous expression of 3′,5′-cyclic AMP (cAMP). This, in turn, triggers a series of consequences and eventually leads to the opening of the cAMP-dependent chloride channel CFTR (cystic fibrosis transmembrane conductance regulator) [75,76,77]. This process is responsible for the pathogenic effects caused by cholera infection, such as rapid fluid loss and rice-water stool [78].
The Ctx B-chain pentamer specifically recognizes the carbohydrate moiety of GM1, particularly GM1a in the a-series of gangliosides, as its receptor (Figure 3) [79]. Structural studies suggest that one Ctx holotoxin could maximally cluster five GM1a molecules on the cell surface [80]. Thus, the Ctx B-chain pentamer has been widely used as a probe for studying ganglioside biology [81]. The biosynthesis of GM1a from LacCer requires three steps (Figure 3): (1) adding CMP-sialic acid to LacCer and generating GM3 by ST3GAL5 (ST3 β-galactoside α-2,3-sialyltransferase 5); (2) adding UDP-N-acetylgalactosamine (UDP-GalNAc) to GM3 and generating GM2 by B4GALNT1 (β-1,4-N-acetylgalactosaminyltransferase 1); and (3) adding UDP-galactose to GM2 and generating GM1a by B3GALT4 (β-1,3-galactosyltransferase 4) [82]. Unlike Gb3, GM1 is widely expressed in human tissues and is involved in multiple essential functions [82].
In 2011, Guimaraes et al. reported a genetic screen for Ctx using a retroviral insertion-based loss-of-function strategy in human haploid KBM7 cells [83]. Since Ctx itself cannot sufficiently kill cells, Guimaraes et al. engineered a lethal chimera toxin by fusing the enzymatic domain of diphtheria toxin (DTA) to the Ctx A chain using the sortase ligation method. DTA is also an ADP-ribosylation enzyme, but it specifically modifies a unique residue called diphthamide in eukaryotic elongation factor 2 (eEF-2), thereby blocking protein synthesis and inducing cell death [84,85]. The screen identified the genes involved in diphthamide biosynthesis that are responsible for DTA, as well as a series of genes involved in GM1a biosynthesis (e.g., UGCG, SLC35A2, B3GALT4, and ST3GAL5). ST3GAL5 is the GM3 synthase, and GM3 is the shared precursor for gangliosides from the a-, b-, and c-series but not from the 0-series (e.g., GM1b, Figure 3) [86]. Guimaraes et al. found that the ST3GAL5-knockout cells were resistant to Ctx. However, Ctx could still bind to a subset of cells (5–10%) at levels comparable to wild-type cells. They speculated that in the absence of ST3GAL5, an alternative ganglioside synthesis pathway for the 0-series can be initiated in a cell cycle-dependent manner. In this case, GM1b acts as an alternative Ctx receptor [83].
In 2014, Gilbert et al. reported another set of genetic screens for the Ctx-DTA chimera toxin using the emerging CRISPRi and CRISPRa approaches [26]. The results of the CRISPRi-based loss-of-function screen suggested that downregulating the genes involved in GM1a biosynthesis (e.g., B3GALT4 and ST3GAL5) leads to the protection of Ctx-DTA. In contrast, downregulating the genes involved in the biosynthesis of other gangliosides, including GM1b (e.g., ST3GAL2), resulted in sensitization to Ctx-DTA. The CRISPRa-based gain-of-function screen yielded results consistent with the CRISPRi screen. In addition, upregulating the genes involved in the biosynthesis of lactosides/neolactosides (e.g., B3GNT5) had protective effects. Both CRISPRi and CRISPRa screens suggested that protection against Ctx-DTA was apparently caused by diverting the shared precursor LacCer away from GM1a synthesis to other branches (Figure 3) [26]. These recent genetic screens expand our understanding of the complex and branched GSL biosynthesis pathways.

3. Factors Involved in Protein Glycosylation

Protein glycosylation is a common form of modification that can occur both co-translationally and post-translationally [87]. There are several types of glycosylation based on the chemistry of which atom from an amino acid residue is attached to the carbohydrate moieties. These types include N-linked (where the nitrogen atom provided by asparagine or arginine is attached), O-linked (where the oxygen atom provided by serine or threonine is attached), C-linked (where the carbon atom provided by tryptophan is attached), and a special type called glypiation (where the C-terminus of the protein is attached by a glycosylphosphatidylinositol (GPI) anchor) (Figure 4).
Glycosylation influences the folding and stability of glycoproteins and is critical for maintaining many cellular functions, such as cell-to-cell communication and adhesion [88]. Congenital disorders of glycosylation (CDG) are a family of human diseases characterized by genetic defects in the glycosylation process, and effective therapies for these disorders are currently lacking [89]. The core reaction of protein glycosylation is initiated in the ER, and the complex structures are synthesized in the Golgi. This enzymatic process requires a large number of enzymes, such as glycosyltransferases, glycosidases, and transporters [90]. Many CDGs have been associated with the loss-of-function mutations identified in these enzymes and the key regulatory factors that control specificity and/or activity [91]. Therefore, a deep understanding of the fundamental glycosylation pathways at the molecular level can help pinpoint therapeutic approaches.
Lectins are a group of proteins that exhibit high specificity in binding to carbohydrate moieties [92]. Many toxins exhibit lectin activity and utilize glycoproteins as their receptors. Examples of such toxins include Clostridioides difficile toxins A (TcdA) and B (TcdB), Escherichia coli subtilase cytotoxin (SubAB), Staphylococcus aureus bi-component pore-forming toxins, insecticidal toxin complexes (Tc toxins), cholesterol-dependent cytolysins (CDCs), pertussis toxin (Ptx), and the plant toxin ricin. These toxins can serve as powerful probes to comprehensively survey protein glycosylation pathways using genome-wide genetic screens. In this section, we focus on recent toxin-based screens for four major glycosylation pathways: N-linked glycosylation, O-linked glycosylation, biosynthesis of sulfated glycosaminoglycans (sGAGs, a special type of O-linked glycosylation), and biosynthesis of GPI anchor (Figure 4 and Table 1).

3.1. Ricin and Related Screens for N-Linked Glycosylation

Ricin, a potential bioterrorism agent, is produced from the seeds of the castor oil plant (Ricinus communis) and has been classified as Biological Select Agents or Toxins (BSATs) by the United States Department of Health and Human Services (HHS) [93]. Ricin is composed of an A chain (32 kDa) and a B chain (34 kDa) connected by an intermolecular disulfide bond [94]. Despite being evolutionarily distinct from Stxs, ricin and Stxs share some common features: (1) Their A chains both function as N-glycosidase and cleave the host ribosomal RNA at the same site, and (2) they both undergo retrograde trafficking pathways and enter the cytoplasm through the ER membrane. The major difference in the mechanism of action between ricin and Stxs is the receptor recognition executed by their respective B chains. Ricin’s B chain is a lectin that broadly binds to the carbohydrate moieties of N-linked glycoproteins containing terminal galactose, or GalNAc [95,96].
Ricin has been screened on eukaryotic cells multiple times using various approaches [26,31,96,97,98,99,100,101]. In 2018, alongside the Stxs screens, Tian et al. also carried out a genome-wide CRISPR-Cas9-mediated screen for ricin in parallel [31]. This screen was conducted in HeLa cells and identified major factors involved in N-linked glycosylation. These factors include the ER-localized glycosyltransferases for producing the dolichol-linked precursor oligosaccharide (e.g., ALG5, ALG6, ALG8, and MOGS), the ER-localized transferase catalyzing the transfer of the precursor oligosaccharide to a protein (e.g., OST4), and the Golgi-localized glycosidases and glycosyltransferases converting the high-mannose precursor oligosaccharide to N-acetylglucosamine (GlcNAc)-containing complex-type glycans (e.g., MAN1A2, MAN2A1, MGAT1, and MGAT2, Figure 4) [31]. The screen also identified key factors in the fucosylation pathway, including cytosolic enzymes for the biosynthesis of GDP-fucose (e.g., TSTA3 and GMDS), and the Golgi-localized GDP-fucose transporter SLC35C1 and fucosyltransferase FUT4 [31]. These results are consistent with the established view that ricin mainly recognizes terminal galactoses on the N-linked glycans as primary receptors. Fucosylation plays a critical role, possibly because transferring fucose to GlcNAc prevents the sialyation of terminal galactoses [96,97,100,101,102].
Tian et al. also identified TMEM165 and TM9SF2 as shared factors between Stxs and ricin, while LAPTM4A is specific to Stxs [31]. They found that TMEM165- and TM9SF2-deficient cells are more resistant to ricin during short-term exposure (20–30 h), but this resistance diminished when the exposure time was extended to 40–50 h. This result suggests that the loss of TMEM165 or TM9SF2 causes a mild reduction in sensitivity to ricin, which is consistent with their indirect roles in regulating N-linked glycosylation.

3.2. TcdA and Related Screens for Biosynthesis of sGAGs

Clostridioides difficile produces two major virulence factors: TcdA and TcdB [103]. Both toxins belong to the large clostridial toxin (LCT) family and consist of four functional domains: (1) an N-terminal enzymatic domain with glucosyltransferase activity; (2) an auto-activation domain; (3) a central domain with both receptor binding and transmembrane delivery activities; and (4) a C-terminal domain with lectin activity and known as combined repetitive oligopeptides (CROPs) [3,104,105,106,107,108,109]. The CROPs domain of TcdA has been shown to broadly interact with the cell surface carbohydrate moieties containing the galactose-β-1,4-GlcNAc motif [110,111,112]. Although the CROPs domains may initiate the attachment of LCT family toxins to the surface of host cells, CROPs-independent receptors have also been identified [32,35,113,114,115].
In 2019, Tao et al. reported a genome-wide CRISPR-Cas9-mediated screen using a truncated form of TcdA without the CROPs domain [33]. The screen identified the low-density lipoprotein receptor (LDLR) as a host factor that mediates TcdA cellular entry through rapid endocytosis and recycling. Surprisingly, the screen also revealed factors in the biosynthesis pathway of sGAG-linked proteoglycan [116]. These factors include the Golgi-localized glycosyltransferases (e.g., XYLT1, XYLT2, B4GALT7, B3GALT6, EXT1, EXT2, EXTL1, EXTL2, and EXTL3), the N-sulfotransferase (e.g., NDST1 and NDST2), the O-sulfotransferase (e.g., HS2ST, HS3ST, and HS6ST1), and the transporter for the activated form of sulfate SLC35B2 (Figure 4) [33]. These results suggest that the sulfation groups in sGAGs may contribute to TcdA binding to cells. Furthermore, the screen identified TMEM165 again, indicating its additional functions in sGAG biosynthesis [33]. Notably, in 2021, another related screen reported by Zhou et al. used a similar strategy for Clostridium novyi alpha-toxin (TcnA, also belonging to the LCT family). They obtained a similar set of sGAG-related genes [117].
In addition to forming and organizing the extracellular matrix, sGAGs exhibit a variety of essential functions, such as signaling transduction and organ development [118]. Defects in sGAG factors have been linked to a large panel of CDGs that occur in multiple tissues, such as Ehlers-Danlos syndrome, Baratela-Scott syndrome, and Langer-Giedion syndrome [119]. These recent toxin-based genetic screens have fully revealed key players in the sGAG biosynthesis pathway, providing not only new biological insights but also potential therapeutic strategies for addressing these disorders.

3.3. TcdB and Related Screens for Biosynthesis of GPI Anchor

TcdB has been shown to recognize two independent receptors, chondroitin sulfate proteoglycan 4 (CSPG4) and frizzled receptors (FZDs), through distinct binding interfaces [113,120,121,122,123,124,125]. Taking advantage of recent sequencing and functional analyses, TcdB subtypes with 3–15% amino acid sequence variations and receptor-binding divergence from the archetype (designated TcdB1) have been identified [126,127]. Intragenic micro-recombination occurring around the receptor-binding regions has been proposed as the driving force for the rapid evolution and diversification of TcdB subtypes [35,127].
In 2022, two back-to-back papers independently reported genome-wide CRISPR-Cas9-mediated screens using subtype TcdB4, and both screens identified tissue factor pathway inhibitor (TFPI) as a novel TcdB receptor [35,114]. TFPI has two major splicing forms: the soluble form TFPIα and the GPI-anchored form TFPIβ [128,129]. TFPIβ is the major form that contributes to TcdB4 cellular binding and entry. Consistently, several well-known enzymes involved in the GPI pathway [130] were also enriched in the screens, such as glycosyltransferases (e.g., PIGA, PIGB, PIGC, PIGM, PIGP, PIGQ, PIGV, PIGX, PIGY, and DPM1), ethanolaminephosphate transferase PIGF, inositol acyltransferase PIGW, and GPI transamidase components (e.g., PIGS, PIGU, and GPAA1) (Figure 4).
In the human proteome, more than 150 proteins use the GPI anchor strategy to display on the cell surface [130]. Defects in GPI biosynthesis have been linked to rare diseases such as hyperphosphatasia with mental retardation syndrome (HPMRS) and paroxysmal nocturnal hemoglobinuria (PNH) [131]. These recent toxin-based genetic screens identified some genes with unknown functions, such as ZNF619, RNF41, PNMA1, C19orf67, and C14orf132 [35,114]. However, whether these genes contribute to GPI biosynthesis remains to be established.

3.4. Other Toxin-Based Screens for Glycosylation Pathways

In 2019, Yamaji et al. reported a CRISPR screen for the Escherichia coli toxin SubAB [132], which is known to utilize terminal sialic acid on glycoproteins as a receptor [133]. Consistently, the screen identified genes involved in both N-linked glycosylation (e.g., MGAT1 and MAN2A1) and O-linked glycosylation (e.g., C1GALT1 and C1GALT1C1), as well as TMEM165 and a predicted Golgi-localized zinc transporter, SLC39A9. Yamaji et al. found that the loss of SLC39A9 reduced both complex-type N-linked glycans, likely through regulating glycosidase MAN2A1, and core 1 O-linked glycans by reducing the key glycosyltransferase C1GALT1. Furthermore, when a key residue (H155) for the predicted Zn2+ transporter activity had been mutated, SLC39A9 lost its ability to regulate glycosylation [132]. Future investigations are required to understand the mechanisms by which SLC39A9 regulates Zn2+ hemostasis and why Zn2+ is involved in glycosylation.
In 2020, Tromp et al. reported two CRISPR screens for the Staphylococcus aureus-produced bi-component pore-forming toxins Panton-Valentine leukocidin (PVL) and γ-haemolysin CB (HlgCB) [134]. These are known to utilize C5AR1 and C5AR2, two G-protein coupled receptors (GPCRs) with post-translational modifications, as receptors [135,136]. The screens identified the known toxins receptor C5AR1 as a positive control and the genes involved in the tyrosine sulfation pathway (e.g., SLC35B2, PAPSS1, and TPST2) as shared host factors between PVL and HlgCB, while the genes involved in the sialyation pathway (e.g., SLC35A1 and CMAS) were unique host factors for HlgCB but not PVL. Tromp et al. further found that sialyation-deficient cells had a lower surface expression level of CXCR2 [134], another GPCR recognized as a receptor for the other two leukotoxins LukED and HlgAB [136,137]. The detailed mechanism by which sialyation regulates surface protein expression remains to be investigated.
In 2020, Drabavicius et al. reported a CRISPR screen in near-haploid HAP1 cells for Streptococcus intermedius-produced CDC intermedilysin (ILY) [138], which has been known to utilize a GPI-anchored protein CD59 as a receptor [139,140]. This screen, along with a similar one in HeLa cells reported by Shahi et al. [141], successfully identified the known ILY receptor CD59 and a whole bunch of host factors in the GPI biosynthesis pathway (e.g., PIGA and PIGB). Drabavicius et al. also identified a panel of genes involved in various cellular processes. These include genes related to N-linked glycosylation (e.g., MGAT1 and MOGS), GSLs biosynthesis (e.g., UGCG and B4GALT5), sGAGs biosynthesis (e.g., EXT2 and B3GALT6), nucleotide sugar metabolism (e.g., GALE, UGP2, and UXS1), as well as TM9SF2 and a subset of genes with unclear functions in ILY intoxication (e.g., C12orf43, C12orf49, TMEM30A, and PDCD10). This screen was so vigorous that it revealed a whole bunch of established factors in multiple glycosylation pathways, possibly due to the enhanced response of haploid HAP1 cells to CRISPR-mediated loss-of-function mutagenesis. Therefore, it is worth paying extra attention to the newly identified factors and their potential roles in glycosylation.
The insecticidal toxin complexes (Tc toxins) produced by entomopathogenic bacteria also exhibit lectin activities [142,143]. Several genetic screens have been conducted to investigate Tc toxins from multiple species using various platforms [144,145]. Through these screens, several genes involved in N-linked glycosylation (e.g., MGAT1 and MAN1A1), GPI biosynthesis (e.g., MPDU1), and sGAG biosynthesis (e.g., EXTL3, SLC35B2, B3GALT6, and Drosophila sgl) have been identified. Notably, TMEM165 and TM9SF2 were identified in a screen for a Tc toxin interacting with sGAGs [144], further confirming their broad involvement in glycosylation.

4. Factors Involved in Membrane Vesicle Trafficking

Many toxins, such as the previously mentioned Stxs, Ctx, ricin, SubAB, pertussis toxin, as well as Pseudomonas aeruginosa exotoxin A (EtA), Salmonella Typhi typhoid toxin, and cytolethal distending toxin (Cdt), exploit retrograde trafficking routes to enter the cytosol. Consistently knocking out or knocking down key factors that regulate trafficking pathways, as well as treatment with small-molecule inhibitors that disrupt the function of the Golgi apparatus (e.g., Brefeldin A) or specifically block trafficking pathways (e.g., Retro-1 and Retro-2 [146]) reduce toxins’ induced toxicities. Therefore, these toxins can be used as probes (different toxins could hijack redundant pathways [99]) to study the membrane vesicle trafficking pathways (Table 1), which are essential for cellular functions, and the defects in these pathways are related to various human diseases [147].

4.1. Screens Using Ricin as a Probe

In 2013, Bassik et al. reported a genome-wide screen for ricin using a short hairpin RNA (shRNA)-based RNAi approach [98]. The screen was primarily conducted using a single-shRNA library (25 shRNAs per gene) to identify the highly confident genes and shRNAs. This was followed by a double-shRNA library based on the results from the primary screen (knockdown two genes simultaneously) to systematically measure the genetic interactions. Bassik et al. identified a set of top genes, including both protective hits (knockdown causing ricin resistance) and sensitizing hits (knockdown causing ricin sensitization) [98]. These top genes were enriched in membrane vesicle trafficking pathways, such as the components of COPII (coat protein complex II, which facilitates ER-Golgi anterograde transport, e.g., SEC24A and SEC24B), TRAPP (transport protein particle, which facilitates ER-Golgi anterograde transport, e.g., TRAPPC8 and TRAPPC11), and GARP (Golgi-associated retrograde protein, which facilitates endosome-Golgi retrograde transport, e.g., VPS53 and VPS54) as protective genes. In contrast, the components of COPI (coat protein complex I, which facilitates endosome-Golgi-ER retrograde transport, e.g., ARCN1 and COPZ1) and Retromer (which facilitates endosome-Golgi retrograde transport, e.g., VPS35 and VPS26A) were identified as sensitizing genes. The finding that losing COPII and TRAPP components causes strong protection against ricin, which was unexpected. This suggested that shutting down ER-budding and anterograde trafficking may cause Golgi dysfunction. By analyzing genetic interactions through the secondary screen, Bassik et al. identified previously poorly characterized genes C4orf41, KIAA1012, and C5orf44 that function as TRAPP interactors, thereby defining two types of TRAPP complexes with distinct compositions and opposite functions in trafficking pathways [98].
A CRISPR-based genetic screen for ricin reported by Tian et al. also revealed several components of the GARP complex (e.g., VPS51, VPS52, VPS53, and VPS54), genes involved in Golgi-ER trafficking (e.g., GOSR1, NAPG, NBAS, STX5, and ARL5B), and the ERAD factors that facilitate toxin crossing the ER membrane (e.g., UBE2G2). Interestingly, in addition to its association with glycosylation pathways, TM9SF2, the shared host factor between ricin and Stxs, was demonstrated to be involved in intracellular vesicular transport. Tian et al. found that TM9SF2-knockout cells exhibited large vacuoles that colocalized with Rab7 within the cytosol and showed abnormal endosomal trafficking across distinct cell types by monitoring the exogenously loaded fluorescently labeled lipids [31]. Consistently, Yamaji et al. showed the disruption of the TGN in TM9SF2-knockout cells and further proposed that TM9SF2 may be required for the retrograde trafficking of glycosyltransferases such as A4GALT [68]. The mechanism underlying the TM9SF2-trafficking axis remains to be established.

4.2. Screens Using Stx as a Probe

In 2017, Selyunin et al. reported a screen for Stxs utilizing a small interfering RNA (siRNA)-based RNAi approach on A4GALT-overexpressing HeLa cells [148]. The screen identified the Golgi-localized protein UNC50 as required for Stx2 endosome-Golgi retrograde trafficking, likely by recruiting the trafficking factor GBF1 (Golgi Brefeldin A resistant guanine nucleotide exchange factor 1) to the Golgi [148]. In the CRISPR screens reported by Sakuma et al., in addition to UNC50, other trafficking factors were identified (e.g., SYS1 and the components of the GARP complex) [68,69]. Particularly, SYS1 (SYS1 Golgi trafficking protein) was previously identified as a host factor for Staphylococcus aureus α-hemolysin (αHL) by regulating the expression of the toxin receptor ADAM10 (ADAM metalloproteinase domain 10) [149]. When SYS1 was knocked out in Vero cells, the TGN showed abnormal morphology and function, resulting in glycosylation defects that made the cells more resistant to Stx [69]. Although UNC50 and SYS1 share some similar features, overexpressing UNC50 in the SYS1 knockout cells failed to compensate for the defects in glycosylation, suggesting that these two factors likely have different functions [69].
In the screens for Stxs reported by Tian et al., the trafficking factor ARCN1 (a COPI component) was identified as a top-ranked hit, whereas neither UNC50 nor SYS1 was identified [31]. This screen recovered fewer trafficking factors, possibly due to the screening strategy relying on long-term toxin treatment (72 h incubation) so that the minor reductions caused by defects in trafficking could be masked. In 2020, Kouzel et al. compared the RNA sequencing profiles between ACHN and Caki-2 cells—two human kidney cell lines that both have Gb3 expression but show different Stx sensitivities [150]. Upon the Stx2 challenge, many trafficking factors showed differential expression, including RAB5A, TRAPPC6B, and YKT6. Notably, neither UNC50 nor SYS1 was identified as differentially expressed genes [150]. The dependence of different trafficking factors on different toxins among various cells or tissues remains to be clarified.

4.3. Screens Using EtA as a Probe

EtA is another toxin that undergoes the retrograde trafficking pathway. Its enzymatic domain acts as an ADP-ribosylation enzyme to deactivate eEF-2, similar to DTA [151]. In 2014, Tafesse et al. reported a genetic screen for EtA using a retrovirus-based mutagenesis strategy in haploid KBM7 cells [152]. The screen successfully identified genes involved in diphthamide biosynthesis (e.g., DPH1, DPH2, and DPH4) corresponding to EtA’s enzymatic activity. A panel of trafficking factors (e.g., components of the GARP complex and the Golgi-ER trafficking factor KDELR1) were also identified. The screen also revealed a new factor, GPR107, a poorly characterized GPCR that had been identified but not characterized in another genetic screen for ricin [101]. Tafesse et al. found that GPR107 is a TGN-localized trafficking factor, and its N-terminal domain, which needs to be activated by furin protease, is critical for retrograde transport [152].
In 2011, Moreau et al. reported two RNAi-based genetic screens for EtA and ricin, respectively [99]. The screens identified a large set of genes involved in membrane vesicle trafficking. This included 65 ricin-specific factors (e.g., components of TRAPP complex), 69 EtA-specific factors (e.g., components of the Retromer and KDELR), and 44 shared factors between two toxins (e.g., components of the GARP complex and STX16). These results demonstrated the genetic complexity of the retrograde trafficking pathway, which is likely not only hijacked by different toxins but also underlies the complexity of cellular membrane-bound compartments [99].

4.4. Screens Using Other Toxins as Probes

In the SubAB screen reported by Yamaji et al., in addition to the genes involved in glycosylation, many trafficking factors were identified (e.g., components of the GARP complex, COG complex, and UNC50) [132]. The screen also revealed KDELR1, KDELR2 (KDEL receptors 1 and 2), and JTB (jumping translocation breakpoint, which were also enriched in a ricin screen [31]). Knocking out these factors did not affect SubAB binding to the cell surface but did suppress the cleavage of the toxin’s intracellular substrate, suggesting their involvement in toxin trafficking [132].
In 2019, Chang et al. reported a CRISPR screen using typhoid toxin as a probe [153]. The screen identified known toxin trafficking factors (e.g., components of the GARP complex, COG (conserved oligomeric Golgi) complex, COPI complex, UNC50, and GPR107) and the ERAD factors (e.g., SEL1L and SYVN1). The screen also revealed TMED2 (transmembrane p24 trafficking protein 2) as a unique Golgi-ER trafficking factor for typhoid toxin, possibly acting as a specific cargo receptor and working together with the COPI complex [153].

5. Factors Involved in Unique Pathways

In 2009, Carette et al. developed a retrovirus-based genetic screen platform that relies on haploid KBM7 cells [18]. This platform has been applied to screen the host factors for diphtheria toxin (DT) and anthrax-diphtheria chimera toxin (anthrax protective antigen (PA) plus anthrax lethal factor N-terminal (LFN) fused with DTA (PA-LFN-DTA)). The screens identified the known toxin receptors (ANTXR2 for PA and HBEGF for DT, respectively) and genes involved in diphthamide biosynthesis (e.g., DPH1, DPH2, and DPH5) corresponding to the enzymatic activity of DTA. A previously uncharacterized gene, WDR85, which is the ortholog of the yeast gene YBR246W, was also identified. Carette et al. found that WDR85 was required for the toxicities but not for the entry of DT, PA-LFN-DTA, or EtA. Furthermore, WDR85 was demonstrated to be involved in diphthamide biosynthesis [18] and has since been renamed DPH7.
By analyzing the genetic interactions through the RNAi-based screen for ricin [98], Bassik et al. found that knocking out the ribosomal protein RPS25 uniquely led to ricin resistance, and RPS25 formed a genetic cluster with the transcription factors ILF2 and ILF3. The strong buffering interactions between RPS25 and ILF2/3 suggest that they may physically interact together to control the translation of certain factors in ricin intoxication. Bassik et al. also identified two previously uncharacterized genes, WDR11 and C17orf75, which form a genetic cluster and interact physically. The WDR11-C17orf75 protein complex may regulate ricin degradation through the autophagy pathway [98].
In 2016, Tao et al. reported a CRISPR screen for TcdB, and the members of the ER membrane protein complex (EMC), such as EMC1, EMC3, EMC4, EMC5, and EMC6, were identified as TcdB host factors. Loss of EMC reduces the expression level of the toxin receptor FZDs [113]. To figure out the full scope of EMC-dependent proteins besides FZDs, Tian et al. carried out a follow-up study using unbiased quantitative proteomic analysis coupled with tandem mass tag labeling and mass spectrometry [154]. Subsets of EMC-dependent and EMC-independent membrane proteins were identified by comparing the membrane protein profiles in wild-type and EMC-knockout cells. Bioinformatic analysis revealed a common feature of EMC-dependent proteins: their transmembrane domains contain polar/charged residues. Introducing or deleting these polar/charged residues can switch the EMC dependency [154].
Recently, Anwar et al. conducted an image-based siRNA screen for anthrax toxin [155]. The screen focused on cell-surface proteins and trafficking factors, utilizing the cleavage of a toxin substrate as a readout. As a result, Anwar et al. identified another TMED member, TMED10, which is essential for anthrax toxin oligomerization on the cell surface. It has been established that TMED10 forms a heterodimer with TMED2. In addition to its role as a cargo receptor in Golgi-ER trafficking, the TMED2/10 complex was demonstrated to facilitate non-vesicular lipid transfer at the ER-Golgi membrane contact site, thereby controlling the formation of plasma membrane lipid nanodomains [155].
Another recently reported CRISPR screen for PVL by Jeon et al. was conducted in differentiated macrophages [156]. Besides the known PVL receptor C5AR1, FBXO11 (F-box protein 11) was also enriched in the screen. Jeon et al. found that FBXO11-knockout cells exhibited reduced transcription of C5AR1 that could be restored by lipopolysaccharide (LPS) priming. FBXO11 was further demonstrated to regulate the inflammasome pathway and the expression of IL-1β through BCL6-dependent and BCL6-independent transcriptional regulations [156].

6. Summary

The toxin-based genetic screens discussed above have been summarized in Table 1. It highlights their screen strategies (e.g., mutagenesis approaches, screen scale, screen formats, and cell models), the identified host factors with known functions, and the newly identified host factors with new functions in fundamental biological pathways. Notably, there is another long list of wonderful studies on toxin–host interactions that have successfully identified toxin–host factors, but they are not discussed here for the following reasons: (1) the identified factors directly interact with toxins, such as receptors; (2) the factors were identified but not functionally characterized; and (3) the factors or pathways were identified by other approaches, such as affinity purification (e.g., pull-down), active labeling (e.g., chemical crosslinking and enzymatic labeling), multi-omics analyses (e.g., proteomics, transcriptomics, lipidomics, and glycomics), and so on.
Table 1. Toxin-based genetic screens are discussed in this review.
Table 1. Toxin-based genetic screens are discussed in this review.
ScreensToxinsScreen Strategy *Identified Host Factors and Functions
Reported by
Tian et al.
[31]
Stx1
Stx2
ricin
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human 5637 and HeLa cells
  • Known genes involved in Gb3 biosynthesis.
  • Known genes involved in N-linked glycosylation and fucosylation pathways.
  • Known genes involved in membrane vesicle trafficking.
  • LAPTM4A: interacts with A4GALT in the Golgi and is required for A4GALT’s enzymatic activity.
  • TMEM165: acts as a Golgi Mn2+ transporter and globally regulates glycosylation.
  • TM9SF2: globally involves in the Golgi glycosylation and membrane vesicle trafficking.
Reported by
Yamaji et al.
[68]
Stx1
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • Known genes involved in Gb3 biosynthesis.
  • Known genes involved in membrane vesicle trafficking.
  • LAPTM4A and TMEM165.
  • TM9SF2: involves in Gb3 biosynthesis likely through A4GALT; and involves in membrane vesicle trafficking.
Reported by
Sakuma et al.
[69]
Stx1
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Green monkey Vero cells **
  • Known genes involved in Gb3 biosynthesis.
  • Known genes involved in membrane vesicle trafficking.
  • LAPTM4A and TM9SF2.
  • SYS1: is required for maintaining Golgi morphology and function.
Reported by
Majumder et al.
[70]
Stx
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • SPTLC1-knockout Human HeLa cells
  • Known genes involved in Gb3 biosynthesis.
  • Known genes involved in membrane vesicle trafficking.
  • LAPTM4A, TMEM165, and TM9SF2.
  • AHR: regulates the expression of Gb3 synthesis factors.
Reported by
Pacheco et al.
[71]
EHEC ***
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HT29 cells
  • Known genes involved in Gb3 biosynthesis.
  • LAPTM4A and TM9SF2.
Reported by
Kono et al.
[72]
Stx
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • SPTLC1-knockout HeLa cells supplementation with S1P
  • Known genes involved in Gb3 biosynthesis.
  • LAPTM4A, TMEM165, and TM9SF2.
  • PLPP3: regulates the uptake of extracellular S1P.
  • SGPP1: dephosphorylates S1P for ceramide synthesis.
Reported by
Selyunin et al.
[148]
Stx1
Stx2
  • RNAi (siRNA)
  • Genome-wide
  • Loss-of-function
  • A4GALT-overexpressing HeLa cells
  • UNC50: is required for Stx2 endosome-Golgi retrograde trafficking.
Reported by
Guimaraes et al.
[83]
Ctx-DTA
  • Retrovirus-based insertional mutagenesis
  • Genome-wide
  • Loss-of-function
  • Human haploid KBM7 cells
  • In the absence of ST3GAL5, 0-series ganglioside synthesis is turned on in a cell cycle-dependent manner.
Reported by
Gilbert et al.
[26]
Ctx-DTA
  • CRISPRi and CRISPRa
  • Genome-wide
  • Loss- and gain-of-function
  • Human K562 cells
  • Upregulation or downregulation of one branch of ganglioside synthesis causes dynamic shifting in other branches.
Reported by
Morgens et al.
[97]
ricin
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human K562 cells
  • Known genes involved in N-linked glycosylation and fucosylation pathways.
Reported by
Bassik et al.
[98]
ricin
  • RNAi (shRNA)
  • Genome-wide single shRNA for the primary screen, paired shRNA for the secondary screen
  • Loss-of-function
  • Human K562 cells
  • Known genes involved in membrane vesicle trafficking.
  • C4orf41, KIAA1012, and C5orf44: as TRAPP interactors and defines two types of TRAPP complexes.
  • RPS25: interacts with transcription factors ILF2 and ILF3.
  • WDR11 and C17orf75: form a complex that may regulate ricin degradation through the autophagy pathway.
Reported by
Tafesse et al.
[152]
EtA
  • Retrovirus-based insertional mutagenesis
  • Genome-wide
  • Loss-of-function
  • Human haploid KBM7 cells
  • Known genes involved in diphthamide biosynthesis.
  • Known genes involved in membrane vesicle trafficking.
  • GPR107: is a TGN-localized GPCR and is critical for retrograde transport.
Reported by
Moreau et al.
[99]
Ricin
EtA
  • RNAi (siRNA)
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • Known genes involved in membrane vesicle trafficking.
  • Shared and unique trafficking pathways for different toxins and membrane-bound compartments.
Reported by
Yamaji et al.
[132]
SubAB
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • Known genes involved in N-linked glycosylation.
  • Known genes involved in O-linked glycosylation.
  • Known genes involved in membrane vesicle trafficking.
  • UNC50 and TMEM165.
  • SLC39A9: acts as a Golgi Zn2+ transporter and globally regulates glycosylation.
  • KDELR1/2 and JTB: involve in trafficking pathway.
Reported by
Chang et al.
[153]
typhoid toxin
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HEK293T cells
  • Known genes involved in membrane vesicle trafficking.
  • Known genes involved in ERAD.
  • UNC50 and GPR107.
  • TMED2: acts as a cargo receptor for Golgi-ER trafficking.
Reported by
Anwar et al.
[155]
Anthrax toxin
  • RNAi (siRNA)
  • 1500 regulatory, trafficking, and cell-surface proteins
  • Loss-of-function
  • Human RPE1 cells
  • TMED10: is essential for anthrax toxin oligomerization on the cell surface.
  • TMED2/10 complex facilitates the non-vesicular transfer of cholesterol and ceramide at the ER-Golgi membrane contact site, thereby controlling the formation of lipid nanodomains.
Reported by
Carette et al.
[18]
DT
LFN-DTA
  • Retrovirus-based insertional mutagenesis
  • Genome-wide
  • Loss-of-function
  • Human haploid KBM7 cells
  • Known genes involved in diphthamide biosynthesis.
  • WDR85: is the ortholog of yeast YBR246W and involves diphthamide biosynthesis (later renamed as DPH7).
Reported by
Tao et al.
[113,154]
TcdB
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • CSPG4 and FZDs: toxin receptors.
  • EMC: facilities membrane protein biosynthesis. The EMC-dependent proteins have polar/charged residues within their transmembrane domains.
Reported by
Tao et al.
[33]
TcdA
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • Known genes involved in sGAGs biosynthesis pathway.
  • LDLR: facilitates toxin uptake.
  • TMEM165.
Reported by
Zhou et al.
[117]
TcnA
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • Known genes involved in sGAGs biosynthesis pathway.
  • LDLR: facilitates toxin uptake.
Reported by
Tian et al.
[35]
TcdB4
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • TFPI: toxin receptor.
  • Known genes involved in the biosynthesis of GPI anchor.
Reported by
Luo et al.
[114]
TcdB4
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • TFPI: toxin receptor.
  • Known genes involved in the biosynthesis of GPI anchor.
Reported by
Tromp et al.
[134]
PVL
HlgCB
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human U937 cells
  • Known genes involved in tyrosine sulfation.
  • Known genes involved in sialyation.
  • Sialyation-deficient cells have lower surface expression level of GPCRs.
Reported by
Jeon et al.
[156]
PVL
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human THP1 macrophages
  • FBXO11: regulates the inflammasome pathway and the expression of IL-1β through transcriptional regulation.
Reported by
Drabavicius et al.
[138]
ILY
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human near-haploid HAP1 cells
  • Known genes involved in the biosynthesis of GPI anchor.
  • Known genes involved in N-linked glycosylation.
  • Known genes involved in GSLs biosynthesis.
  • Known genes involved in sGAGs biosynthesis pathway.
  • Known genes involved in nucleotide sugar metabolism.
  • TM9SF2.
Reported by
Shahi et al.
[141]
ILY
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • Known genes involved in the biosynthesis of GPI anchor.
Reported by
Virreira Winter et al. [149]
αHL
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human U937 cells
  • SYS1, ARFRP1, and TSPAN14: regulate the surface expression of toxin receptor ADAM10.
Reported by
Song et al.
[144]
Tc toxins
  • CRISPR-Cas9
  • Genome-wide
  • Loss-of-function
  • Human HeLa cells
  • Known genes involved in the biosynthesis of GPI anchor.
  • Known genes involved in N-linked glycosylation.
  • Known genes involved in sGAGs biosynthesis pathway.
  • TMEM165 and TM9SF2.
* Screen strategies encompass various technical considerations, such as screen scale (partial genome or genome-wide), cell model, methods of generating mutations, screen format (loss-of-function or gain-of-function), and some special settings. ** The loss-of-function approach involved using a library targeting the human genome in Vero cells derived from green monkeys. *** The EHEC strain used in this screen expresses both T3SS and Stx.

7. Perspectives

The journey of studying bacterial toxins began in the 19th century. In 1884, Robert Koch pointed out that Vibrio cholerae induces disease through a secreted “poison,” which was later known as Ctx after more than half a century. In 1883, Corynebacterium diphtheriae was identified as the causative agent of diphtheria. In 1888, Émile Roux and Alexandre Yersin discovered and defined the first bacterial toxin, diphtheria toxin (DT), from the supernatant of Corynebacterium diphtheriae culture. This discovery demonstrated that bacteria could produce a particular substance that acts as the disease-causing agent, and the substance was named as bacterial toxin. In 1901, a toxin biologist, Emil von Behring, was awarded the first Nobel Prize in Physiology or Medicine for his work on developing antiserum against DT and tetanus toxin. Since then, more than 300 toxins have been discovered [157].
Significant progress has been made in terms of research on toxin biology and toxin–host interactions. On the toxin side, the biochemical and structural features of toxin have been elucidated, while on the host side, the cellular mechanism of their actions has been disclosed. These achievements have provided insights into how these potential bioterrorism agents have become the most poisonous substances in the world, with BoNT and TeNT being claimed as the most potent toxins, ranked No. 1 and No. 2, respectively [158]. On the other hand, these naturally evolved properties of toxins provide a powerful toolbox for engineering and developing toxin-based platforms. These platforms can be tailored to target specific cells or tissues, enable efficient intracellular delivery [159], and modulate signaling pathways.
This review specifically focuses on the combination of toxin biology and recently advanced genetic screen technologies—toxins serve as specific and robust probes, while genetic screens provide systematic and unbiased surveys. Therefore, toxin-based genetic screens can reveal a comprehensive list of host factors involved in toxin intoxication. Particularly, host factors that indirectly interact with toxins may play important roles in fundamental biological pathways and eventually benefit toxins. These host factors may be involved in controlling the biosynthesis of toxin receptors, facilitating toxin trafficking among membrane-bound organelles, or being required for the toxin’s enzymatic activity.
The successful identification of these factors highly relies on the employed screen strategies. For example, RNAi-based screens for ricin identified many trafficking factors, whereas CRISPR-based screens for ricin additionally revealed N-linked glycosylation and fucosylation pathways. Similarly, multiple CRISPR-based screens for Stx consistently identified LAPTM4A and TM9SF2, which had not been recognized by previous RNAi-based screens. Furthermore, the PVL screen on a regular cell line only provided information on the receptor-related pathways, whereas the same screen conducted in macrophages discovered regulators in the inflammasome pathway. Moreover, some minor conditions, such as toxin concentration and treatment time, can also have an impact. Future investigations in this area are required, involving more screens using various strategies and delving into the molecular mechanisms of these newly identified factors and pathways.

Author Contributions

Conceptualization, S.T.; writing—original draft preparation, S.T.; writing—review and editing, S.T. and N.Z.; visualization, S.T.; supervision, S.T. All authors have read and agreed to the published version of the manuscript.

Funding

This study was partially supported in part by grants from the National Institutes of Health (NIH) (R01NS080833, R01NS117626, R01AI132387, R21NS106159, R01AI139087, and R01AI158503).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Min Dong for the helpful discussion and financial support.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

StxShiga toxin
Ctxcholera toxin
DTdiphtheria toxin
DTAenzymatic domain of diphtheria toxin
PAanthrax protective antigen
LFNanthrax lethal factor N-terminal
EtAPseudomonas aeruginosa exotoxin A
LCTlarge clostridial toxin
TcdAClostridioides difficile toxin A
TcdBClostridioides difficile toxin B
TcnAClostridium novyi alpha-toxin
CROPscombined repetitive oligopeptides
SubABEscherichia coli subtilase cytotoxin
CDCcholesterol-dependent cytolysin
Tc toxininsecticidal toxin complex
αHLStaphylococcus aureus α-hemolysin
PVLStaphylococcus aureus Panton-Valentine leucocidin
HlgCBStaphylococcus aureus γ-haemolysin CB
ILYStreptococcus intermedius intermedilysin
Cdtcytolethal distending toxin
T3SStype III secretion system
LPSlipopolysaccharides
EHECenterohemorrhagic Escherichia coli
RNAiRNA interference
CRISPRclustered regularly interspaced short palindromic repeat
CRISPRiCRISPR repression
CRISPRaCRISPR activation
sgRNAsingle guide RNA
shRNAshort hairpin RNA
siRNAsmall interfering RNA
cDNAcomplementary DNA
cAMP3′,5′-cyclic AMP
GalNAcN-acetylgalactosamine
GlcNAcN-acetylglucosamine
GSLglycosphingolipid
GlcCerglucosylceramide
LacCerlactosylceramide
Gb3globotriaosylceramide (also known as CD77)
GM1monosialotetrahexosylganglioside
S1Psphingosine-1-phosphate
GPIglycosylphosphatidylinositol
GAGglycosaminoglycan
sGAGsulfated glycosaminoglycan
ERendoplasmic reticulum
TGNtrans-Golgi network
ERADER-associated protein degradation
GPCRG-protein coupled receptor
COPIcoat protein complex I
COPIIcoat protein complex II
TRAPPtransport protein particle
GARPGolgi-associated retrograde protein
COGconserved oligomeric Golgi complex
EMCER membrane protein complex
UGCGUDP-glucose ceramide glucosyltransferase
B4GALT5β-1,4-galactosyltransferase 5
SLC35A2solute carrier family 35 member A2
A4GALTα-1,4-galactosyltransferase (also known as Gb3 synthetase)
SPTSSAserine palmitoyltransferase small subunit A
SPTLC1serine palmitoyltransferase long chain base subunit 1
UGP2UDP-glucose pyrophosphorylase 2
SPPL3Signal peptide peptidase-like 3
LAPTM4Alysosomal-associated protein transmembrane 4 A
LAPTM4Blysosomal-associated protein transmembrane 4 B
TMEM165transmembrane protein 165
TM9SF2transmembrane 9 superfamily member 2
AHRaryl hydrocarbon receptor
PLPP3phospholipid phosphatase 3 (also known as PPAP2B)
SGPP1S1P phosphatase 1
CFTRcystic fibrosis transmembrane conductance regulator
ST3GAL5ST3 β-galactoside α-2,3-sialyltransferase 5
B4GALNT1β-1,4-N-acetylgalactosaminyltransferase 1
B3GALT4β-1,3-galactosyltransferase 4
eEF-2eukaryotic elongation factor 2
CSPG4chondroitin sulfate proteoglycan 4
FZDsfrizzled receptors
SLC39A9solute carrier family 39 member A9
GBF1Golgi Brefeldin A resistant guanine nucleotide exchange factor 1
ADAM10 ADAM metalloproteinase domain 10
KDELR1KDEL receptors 1
KDELR2KDEL receptors 2
JTBjumping translocation breakpoint
TMED2transmembrane p24 trafficking protein 2
FBXO11F-box protein 11
D+HUSpost-diarrheal hemolytic uremic syndrome
CDGcongenital disorders of glycosylation
HPMRShyperphosphatasia with mental retardation syndrome
PNHparoxysmal nocturnal hemoglobinuria

References

  1. Ala’Aldeen, D.A.A.; Wooldridge, K.G. Bacterial pathogenicity. In Medical Microbiology; Elsevier: Amsterdam, The Netherlands, 2012; pp. 156–167. [Google Scholar] [CrossRef]
  2. Barbieri, J.T. Exotoxins. In Encyclopedia of Microbiology; Elsevier: Amsterdam, The Netherlands, 2009; pp. 355–364. [Google Scholar] [CrossRef]
  3. Orrell, K.E.; Melnyk, R.A. Large Clostridial Toxins: Mechanisms and Roles in Disease. Microbiol. Mol. Biol. Rev. 2021, 85, e0006421. [Google Scholar] [CrossRef]
  4. 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]
  5. Odumosu, O.; Nicholas, D.; Yano, H.; Langridge, W. AB toxins: A paradigm switch from deadly to desirable. Toxins 2010, 2, 1612–1645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Beddoe, T.; Paton, A.W.; Le Nours, J.; Rossjohn, J.; Paton, J.C. Structure, biological functions and applications of the AB5 toxins. Trends Biochem. Sci. 2010, 35, 411–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Pons, B.J.; Vignard, J.; Mirey, G. Cytolethal Distending Toxin Subunit B: A Review of Structure-Function Relationship. Toxins 2019, 11, 595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Fowler, C.C.; Chang, S.J.; Gao, X.; Geiger, T.; Stack, G.; Galan, J.E. Emerging insights into the biology of typhoid toxin. Curr. Opin. Microbiol. 2017, 35, 70–77. [Google Scholar] [CrossRef] [Green Version]
  9. Dong, M.; Masuyer, G.; Stenmark, P. Botulinum and Tetanus Neurotoxins. Annu. Rev. Biochem. 2019, 88, 811–837. [Google Scholar] [CrossRef]
  10. Moresco, E.M.; Li, X.; Beutler, B. Going forward with genetics: Recent technological advances and forward genetics in mice. Am. J. Pathol. 2013, 182, 1462–1473. [Google Scholar] [CrossRef] [Green Version]
  11. Grimm, S. The art and design of genetic screens: Mammalian culture cells. Nat. Rev. Genet. 2004, 5, 179–189. [Google Scholar] [CrossRef]
  12. Strachan, T.; Read, A.P.; Strachan, T. Human Molecular Genetics, 2nd ed.; BIOS Scientific Publishers Ltd.: Oxford, UK, 1999; p. xxiii. 576p. [Google Scholar]
  13. Collaboration, O.R. The ORFeome Collaboration: A genome-scale human ORF-clone resource. Nat. Methods 2016, 13, 191–192. [Google Scholar] [CrossRef]
  14. Yang, X.; Boehm, J.S.; Yang, X.; Salehi-Ashtiani, K.; Hao, T.; Shen, Y.; Lubonja, R.; Thomas, S.R.; Alkan, O.; Bhimdi, T.; et al. A public genome-scale lentiviral expression library of human ORFs. Nat. Methods 2011, 8, 659–661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Naglich, J.G.; Metherall, J.E.; Russell, D.W.; Eidels, L. Expression cloning of a diphtheria toxin receptor: Identity with a heparin-binding EGF-like growth factor precursor. Cell 1992, 69, 1051–1061. [Google Scholar] [CrossRef] [PubMed]
  16. Berns, K.; Hijmans, E.M.; Mullenders, J.; Brummelkamp, T.R.; Velds, A.; Heimerikx, M.; Kerkhoven, R.M.; Madiredjo, M.; Nijkamp, W.; Weigelt, B.; et al. A large-scale RNAi screen in human cells identifies new components of the p53 pathway. Nature 2004, 428, 431–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Paddison, P.J.; Silva, J.M.; Conklin, D.S.; Schlabach, M.; Li, M.; Aruleba, S.; Balija, V.; O’Shaughnessy, A.; Gnoj, L.; Scobie, K.; et al. A resource for large-scale RNA-interference-based screens in mammals. Nature 2004, 428, 427–431. [Google Scholar] [CrossRef]
  18. Carette, J.E.; Guimaraes, C.P.; Varadarajan, M.; Park, A.S.; Wuethrich, I.; Godarova, A.; Kotecki, M.; Cochran, B.H.; Spooner, E.; Ploegh, H.L.; et al. Haploid genetic screens in human cells identify host factors used by pathogens. Science 2009, 326, 1231–1235. [Google Scholar] [CrossRef] [Green Version]
  19. Carette, J.E.; Guimaraes, C.P.; Wuethrich, I.; Blomen, V.A.; Varadarajan, M.; Sun, C.; Bell, G.; Yuan, B.; Muellner, M.K.; Nijman, S.M.; et al. Global gene disruption in human cells to assign genes to phenotypes by deep sequencing. Nat. Biotechnol. 2011, 29, 542–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Shalem, O.; Sanjana, N.E.; Hartenian, E.; Shi, X.; Scott, D.A.; Mikkelson, T.; Heckl, D.; Ebert, B.L.; Root, D.E.; Doench, J.G.; et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 2014, 343, 84–87. [Google Scholar] [CrossRef] [Green Version]
  21. Wang, T.; Wei, J.J.; Sabatini, D.M.; Lander, E.S. Genetic screens in human cells using the CRISPR-Cas9 system. Science 2014, 343, 80–84. [Google Scholar] [CrossRef] [Green Version]
  22. Shalem, O.; Sanjana, N.E.; Zhang, F. High-throughput functional genomics using CRISPR-Cas9. Nat. Rev. Genet. 2015, 16, 299–311. [Google Scholar] [CrossRef] [Green Version]
  23. Puschnik, A.S.; Majzoub, K.; Ooi, Y.S.; Carette, J.E. A CRISPR toolbox to study virus-host interactions. Nat. Rev. Microbiol. 2017, 15, 351–364. [Google Scholar] [CrossRef] [PubMed]
  24. Gilbert, L.A.; Larson, M.H.; Morsut, L.; Liu, Z.; Brar, G.A.; Torres, S.E.; Stern-Ginossar, N.; Brandman, O.; Whitehead, E.H.; Doudna, J.A.; et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell 2013, 154, 442–451. [Google Scholar] [CrossRef] [Green Version]
  25. Qi, L.S.; Larson, M.H.; Gilbert, L.A.; Doudna, J.A.; Weissman, J.S.; Arkin, A.P.; Lim, W.A. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell 2013, 152, 1173–1183. [Google Scholar] [CrossRef] [Green Version]
  26. Gilbert, L.A.; Horlbeck, M.A.; Adamson, B.; Villalta, J.E.; Chen, Y.; Whitehead, E.H.; Guimaraes, C.; Panning, B.; Ploegh, H.L.; Bassik, M.C.; et al. Genome-Scale CRISPR-Mediated Control of Gene Repression and Activation. Cell 2014, 159, 647–661. [Google Scholar] [CrossRef] [Green Version]
  27. Boutros, M.; Ahringer, J. The art and design of genetic screens: RNA interference. Nat. Rev. Genet. 2008, 9, 554–566. [Google Scholar] [CrossRef] [PubMed]
  28. Goncalves, J.; Moreira, E.; Sequeira, I.J.; Rodrigues, A.S.; Rueff, J.; Bras, A. Integration of HIV in the Human Genome: Which Sites Are Preferential? A Genetic and Statistical Assessment. Int. J. Genom. 2016, 2016, 2168590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Konermann, S.; Brigham, M.D.; Trevino, A.E.; Joung, J.; Abudayyeh, O.O.; Barcena, C.; Hsu, P.D.; Habib, N.; Gootenberg, J.S.; Nishimasu, H.; et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 2015, 517, 583–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Fu, Y.; Foden, J.A.; Khayter, C.; Maeder, M.L.; Reyon, D.; Joung, J.K.; Sander, J.D. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat. Biotechnol. 2013, 31, 822–826. [Google Scholar] [CrossRef] [Green Version]
  31. Tian, S.; Muneeruddin, K.; Choi, M.Y.; Tao, L.; Bhuiyan, R.H.; Ohmi, Y.; Furukawa, K.; Furukawa, K.; Boland, S.; Shaffer, S.A.; et al. Genome-wide CRISPR screens for Shiga toxins and ricin reveal Golgi proteins critical for glycosylation. PLoS Biol. 2018, 16, e2006951. [Google Scholar] [CrossRef] [Green Version]
  32. Tian, S.; Liu, Y.; Wu, H.; Liu, H.; Zeng, J.; Choi, M.Y.; Chen, H.; Gerhard, R.; Dong, M. Genome-Wide CRISPR Screen Identifies Semaphorin 6A and 6B as Receptors for Paeniclostridium sordellii Toxin TcsL. Cell Host Microbe 2020, 27, 782–792.e7. [Google Scholar] [CrossRef]
  33. Tao, L.; Tian, S.; Zhang, J.; Liu, Z.; Robinson-McCarthy, L.; Miyashita, S.I.; Breault, D.T.; Gerhard, R.; Oottamasathien, S.; Whelan, S.P.J.; et al. Sulfated glycosaminoglycans and low-density lipoprotein receptor contribute to Clostridium difficile toxin A entry into cells. Nat. Microbiol. 2019, 4, 1760–1769. [Google Scholar] [CrossRef]
  34. Xiong, X.; Tian, S.; Yang, P.; Lebreton, F.; Bao, H.; Sheng, K.; Yin, L.; Chen, P.; Zhang, J.; Qi, W.; et al. Emerging enterococcus pore-forming toxins with MHC/HLA-I as receptors. Cell 2022, 185, 1157–1171.e22. [Google Scholar] [CrossRef] [PubMed]
  35. Tian, S.; Xiong, X.; Zeng, J.; Wang, S.; Tremblay, B.J.; Chen, P.; Chen, B.; Liu, M.; Chen, P.; Sheng, K.; et al. Identification of TFPI as a receptor reveals recombination-driven receptor switching in Clostridioides difficile toxin B variants. Nat. Commun. 2022, 13, 6786. [Google Scholar] [CrossRef]
  36. Holthuis, J.C.; Menon, A.K. Lipid landscapes and pipelines in membrane homeostasis. Nature 2014, 510, 48–57. [Google Scholar] [CrossRef] [PubMed]
  37. Yamaji, T.; Hanada, K. Sphingolipid metabolism and interorganellar transport: Localization of sphingolipid enzymes and lipid transfer proteins. Traffic 2015, 16, 101–122. [Google Scholar] [CrossRef] [PubMed]
  38. Kolter, T.; Sandhoff, K. Sphingolipid metabolism diseases. Biochim. Biophys. Acta 2006, 1758, 2057–2079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Green, C.D.; Maceyka, M.; Cowart, L.A.; Spiegel, S. Sphingolipids in metabolic disease: The good, the bad, and the unknown. Cell Metab. 2021, 33, 1293–1306. [Google Scholar] [CrossRef]
  40. Chester, M.A. IUPAC-IUB Joint Commission on Biochemical Nomenclature (JCBN). Nomenclature of glycolipids—Recommendations 1997. Eur. J. Biochem. 1998, 257, 293–298. [Google Scholar] [CrossRef]
  41. Qasba, P.K.; Ramakrishnan, B. Letter to the Glyco-Forum: Catalytic domains of glycosyltransferases with ‘add-on’ domains. Glycobiology 2007, 17, 7G–9G. [Google Scholar] [CrossRef]
  42. Kitatani, K.; Idkowiak-Baldys, J.; Hannun, Y.A. The sphingolipid salvage pathway in ceramide metabolism and signaling. Cell. Signal. 2008, 20, 1010–1018. [Google Scholar] [CrossRef] [Green Version]
  43. Merrill, A.H., Jr. Sphingolipid and glycosphingolipid metabolic pathways in the era of sphingolipidomics. Chem. Rev. 2011, 111, 6387–6422. [Google Scholar] [CrossRef]
  44. Geny, B.; Popoff, M.R. Bacterial protein toxins and lipids: Role in toxin targeting and activity. Biol. Cell 2006, 98, 633–651. [Google Scholar] [CrossRef] [PubMed]
  45. Johannes, L.; Romer, W. Shiga toxins—From cell biology to biomedical applications. Nat. Rev. Microbiol. 2010, 8, 105–116. [Google Scholar] [CrossRef] [PubMed]
  46. Bergan, J.; Dyve Lingelem, A.B.; Simm, R.; Skotland, T.; Sandvig, K. Shiga toxins. Toxicon 2012, 60, 1085–1107. [Google Scholar] [CrossRef]
  47. Lingwood, C.A. Verotoxins and their glycolipid receptors. Adv. Lipid Res. 1993, 25, 189–211. [Google Scholar] [PubMed]
  48. Ling, H.; Boodhoo, A.; Hazes, B.; Cummings, M.D.; Armstrong, G.D.; Brunton, J.L.; Read, R.J. Structure of the shiga-like toxin I B-pentamer complexed with an analogue of its receptor Gb3. Biochemistry 1998, 37, 1777–1788. [Google Scholar] [CrossRef] [PubMed]
  49. Kojima, Y.; Fukumoto, S.; Furukawa, K.; Okajima, T.; Wiels, J.; Yokoyama, K.; Suzuki, Y.; Urano, T.; Ohta, M.; Furukawa, K. Molecular cloning of globotriaosylceramide/CD77 synthase, a glycosyltransferase that initiates the synthesis of globo series glycosphingolipids. J. Biol. Chem. 2000, 275, 15152–15156. [Google Scholar] [CrossRef] [Green Version]
  50. Keusch, J.J.; Manzella, S.M.; Nyame, K.A.; Cummings, R.D.; Baenziger, J.U. Cloning of Gb3 synthase, the key enzyme in globo-series glycosphingolipid synthesis, predicts a family of alpha 1, 4-glycosyltransferases conserved in plants, insects, and mammals. J. Biol. Chem. 2000, 275, 25315–25321. [Google Scholar] [CrossRef] [Green Version]
  51. Okuda, T.; Tokuda, N.; Numata, S.; Ito, M.; Ohta, M.; Kawamura, K.; Wiels, J.; Urano, T.; Tajima, O.; Furukawa, K.; et al. Targeted disruption of Gb3/CD77 synthase gene resulted in the complete deletion of globo-series glycosphingolipids and loss of sensitivity to verotoxins. J. Biol. Chem. 2006, 281, 10230–10235. [Google Scholar] [CrossRef] [Green Version]
  52. Kannagi, R.; Fukuda, M.N.; Hakomori, S. A new glycolipid antigen isolated from human erythrocyte membranes reacting with antibodies directed to globo-N-tetraosylceramide (globoside). J. Biol. Chem. 1982, 257, 4438–4442. [Google Scholar] [CrossRef]
  53. Gregory, C.D.; Dive, C.; Henderson, S.; Smith, C.A.; Williams, G.T.; Gordon, J.; Rickinson, A.B. Activation of Epstein-Barr virus latent genes protects human B cells from death by apoptosis. Nature 1991, 349, 612–614. [Google Scholar] [CrossRef]
  54. Liu, Y.; Tian, S.; Thaker, H.; Dong, M. Shiga Toxins: An Update on Host Factors and Biomedical Applications. Toxins 2021, 13, 222. [Google Scholar] [CrossRef] [PubMed]
  55. Obrig, T.G.; Karpman, D. Shiga toxin pathogenesis: Kidney complications and renal failure. Curr. Top. Microbiol. Immunol. 2012, 357, 105–136. [Google Scholar] [CrossRef] [Green Version]
  56. Masson, C.; Cisse, I.; Simon, V.; Insalaco, P.; Audran, M. Fabry disease: A review. Joint Bone Spine 2004, 71, 381–383. [Google Scholar] [CrossRef] [PubMed]
  57. Mahmud, H.M. Fabry’s disease—A comprehensive review on pathogenesis, diagnosis and treatment. J. Pak. Med. Assoc. 2014, 64, 189–194. [Google Scholar] [PubMed]
  58. Zarate, Y.A.; Hopkin, R.J. Fabry’s disease. Lancet 2008, 372, 1427–1435. [Google Scholar] [CrossRef] [PubMed]
  59. Voss, M.; Kunzel, U.; Higel, F.; Kuhn, P.H.; Colombo, A.; Fukumori, A.; Haug-Kroper, M.; Klier, B.; Grammer, G.; Seidl, A.; et al. Shedding of glycan-modifying enzymes by signal peptide peptidase-like 3 (SPPL3) regulates cellular N-glycosylation. EMBO J. 2014, 33, 2890–2905. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Kuhn, P.H.; Voss, M.; Haug-Kroper, M.; Schroder, B.; Schepers, U.; Brase, S.; Haass, C.; Lichtenthaler, S.F.; Fluhrer, R. Secretome analysis identifies novel signal Peptide peptidase-like 3 (Sppl3) substrates and reveals a role of Sppl3 in multiple Golgi glycosylation pathways. Mol. Cell. Proteom. 2015, 14, 1584–1598. [Google Scholar] [CrossRef] [Green Version]
  61. Hogue, D.L.; Ellison, M.J.; Young, J.D.; Cass, C.E. Identification of a novel membrane transporter associated with intracellular membranes by phenotypic complementation in the yeast Saccharomyces cerevisiae. J. Biol. Chem. 1996, 271, 9801–9808. [Google Scholar] [CrossRef] [Green Version]
  62. Cabrita, M.A.; Hobman, T.C.; Hogue, D.L.; King, K.M.; Cass, C.E. Mouse transporter protein, a membrane protein that regulates cellular multidrug resistance, is localized to lysosomes. Cancer Res. 1999, 59, 4890–4897. [Google Scholar]
  63. Potelle, S.; Morelle, W.; Dulary, E.; Duvet, S.; Vicogne, D.; Spriet, C.; Krzewinski-Recchi, M.A.; Morsomme, P.; Jaeken, J.; Matthijs, G.; et al. Glycosylation abnormalities in Gdt1p/TMEM165 deficient cells result from a defect in Golgi manganese homeostasis. Hum. Mol. Genet. 2016, 25, 1489–1500. [Google Scholar] [CrossRef] [Green Version]
  64. Foulquier, F.; Amyere, M.; Jaeken, J.; Zeevaert, R.; Schollen, E.; Race, V.; Bammens, R.; Morelle, W.; Rosnoblet, C.; Legrand, D.; et al. TMEM165 deficiency causes a congenital disorder of glycosylation. Am. J. Hum. Genet. 2012, 91, 15–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Dulary, E.; Potelle, S.; Legrand, D.; Foulquier, F. TMEM165 deficiencies in Congenital Disorders of Glycosylation type II (CDG-II): Clues and evidences for roles of the protein in Golgi functions and ion homeostasis. Tissue Cell 2017, 49, 150–156. [Google Scholar] [CrossRef]
  66. Schimmoller, F.; Diaz, E.; Muhlbauer, B.; Pfeffer, S.R. Characterization of a 76 kDa endosomal, multispanning membrane protein that is highly conserved throughout evolution. Gene 1998, 216, 311–318. [Google Scholar] [CrossRef]
  67. Tanaka, A.; Tumkosit, U.; Nakamura, S.; Motooka, D.; Kishishita, N.; Priengprom, T.; Sa-Ngasang, A.; Kinoshita, T.; Takeda, N.; Maeda, Y. Genome-Wide Screening Uncovers the Significance of N-Sulfation of Heparan Sulfate as a Host Cell Factor for Chikungunya Virus Infection. J. Virol. 2017, 91, e00432-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Yamaji, T.; Sekizuka, T.; Tachida, Y.; Sakuma, C.; Morimoto, K.; Kuroda, M.; Hanada, K. A CRISPR Screen Identifies LAPTM4A and TM9SF Proteins as Glycolipid-Regulating Factors. iScience 2019, 11, 409–424. [Google Scholar] [CrossRef] [Green Version]
  69. Sakuma, C.; Sekizuka, T.; Kuroda, M.; Hanada, K.; Yamaji, T. Identification of SYS1 as a Host Factor Required for Shiga Toxin-Mediated Cytotoxicity in Vero Cells. Int. J. Mol. Sci. 2021, 22, 4936. [Google Scholar] [CrossRef]
  70. Majumder, S.; Kono, M.; Lee, Y.T.; Byrnes, C.; Li, C.; Tuymetova, G.; Proia, R.L. A genome-wide CRISPR/Cas9 screen reveals that the aryl hydrocarbon receptor stimulates sphingolipid levels. J. Biol. Chem. 2020, 295, 4341–4349. [Google Scholar] [CrossRef] [Green Version]
  71. Pacheco, A.R.; Lazarus, J.E.; Sit, B.; Schmieder, S.; Lencer, W.I.; Blondel, C.J.; Doench, J.G.; Davis, B.M.; Waldor, M.K. CRISPR Screen Reveals that EHEC’s T3SS and Shiga Toxin Rely on Shared Host Factors for Infection. mBio 2018, 9, e01003-18. [Google Scholar] [CrossRef] [Green Version]
  72. Kono, M.; Hoachlander-Hobby, L.E.; Majumder, S.; Schwartz, R.; Byrnes, C.; Zhu, H.; Proia, R.L. Identification of two lipid phosphatases that regulate sphingosine-1-phosphate cellular uptake and recycling. J. Lipid Res. 2022, 63, 100225. [Google Scholar] [CrossRef] [PubMed]
  73. De, S.N.; Sarkar, J.K.; Tribedi, B.P. An experimental study of the action of cholera toxin. J. Pathol. Bacteriol. 1951, 63, 707–717. [Google Scholar] [CrossRef]
  74. Lencer, W.I.; Tsai, B. The intracellular voyage of cholera toxin: Going retro. Trends Biochem. Sci. 2003, 28, 639–645. [Google Scholar] [CrossRef]
  75. Schafer, D.E.; Lust, W.D.; Sircar, B.; Goldberg, N.D. Elevated concentration of adenosine 3′:5′-cyclic monophosphate in intestinal mucosa after treatment with cholera toxin. Proc. Natl. Acad. Sci. USA 1970, 67, 851–856. [Google Scholar] [CrossRef] [PubMed]
  76. Kassis, S.; Hagmann, J.; Fishman, P.H.; Chang, P.P.; Moss, J. Mechanism of action of cholera toxin on intact cells. Generation of A1 peptide and activation of adenylate cyclase. J. Biol. Chem. 1982, 257, 12148–12152. [Google Scholar] [CrossRef] [PubMed]
  77. Kahn, R.A.; Gilman, A.G. ADP-ribosylation of Gs promotes the dissociation of its alpha and beta subunits. J. Biol. Chem. 1984, 259, 6235–6240. [Google Scholar] [CrossRef]
  78. Bharati, K.; Ganguly, N.K. Cholera toxin: A paradigm of a multifunctional protein. Indian J. Med. Res. 2011, 133, 179–187. [Google Scholar]
  79. Heyningen, S.V. Cholera toxin: Interaction of subunits with ganglioside GM1. Science 1974, 183, 656–657. [Google Scholar] [CrossRef] [PubMed]
  80. Merritt, E.A.; Sarfaty, S.; van den Akker, F.; L’Hoir, C.; Martial, J.A.; Hol, W.G. Crystal structure of cholera toxin B-pentamer bound to receptor GM1 pentasaccharide. Protein Sci. 1994, 3, 166–175. [Google Scholar] [CrossRef] [Green Version]
  81. Kenworthy, A.K.; Schmieder, S.S.; Raghunathan, K.; Tiwari, A.; Wang, T.; Kelly, C.V.; Lencer, W.I. Cholera Toxin as a Probe for Membrane Biology. Toxins 2021, 13, 543. [Google Scholar] [CrossRef]
  82. Ledeen, R.W.; Wu, G. The multi-tasked life of GM1 ganglioside, a true factotum of nature. Trends Biochem. Sci. 2015, 40, 407–418. [Google Scholar] [CrossRef]
  83. Guimaraes, C.P.; Carette, J.E.; Varadarajan, M.; Antos, J.; Popp, M.W.; Spooner, E.; Brummelkamp, T.R.; Ploegh, H.L. Identification of host cell factors required for intoxication through use of modified cholera toxin. J. Cell Biol. 2011, 195, 751–764. [Google Scholar] [CrossRef]
  84. Collier, R.J. Understanding the mode of action of diphtheria toxin: A perspective on progress during the 20th century. Toxicon 2001, 39, 1793–1803. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, S.; Milne, G.T.; Kuremsky, J.G.; Fink, G.R.; Leppla, S.H. Identification of the proteins required for biosynthesis of diphthamide, the target of bacterial ADP-ribosylating toxins on translation elongation factor 2. Mol. Cell. Biol. 2004, 24, 9487–9497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Yamashita, T.; Hashiramoto, A.; Haluzik, M.; Mizukami, H.; Beck, S.; Norton, A.; Kono, M.; Tsuji, S.; Daniotti, J.L.; Werth, N.; et al. Enhanced insulin sensitivity in mice lacking ganglioside GM3. Proc. Natl. Acad. Sci. USA 2003, 100, 3445–3449. [Google Scholar] [CrossRef]
  87. Stanley, P. Golgi glycosylation. Cold Spring Harb. Perspect. Biol. 2011, 3, a005199. [Google Scholar] [CrossRef] [PubMed]
  88. Varki, A.; Cummings, R.D.; Esko, J.D.; Freeze, H.H.; Stanley, P.; Bertozzi, C.R.; Hart, G.W.; Etzler, M.E. (Eds.) Essentials of Glycobiology, 2nd ed.; Cold Spring Harbor (NY): New York, NY, USA, 2009. [Google Scholar]
  89. Freeze, H.H.; Chong, J.X.; Bamshad, M.J.; Ng, B.G. Solving glycosylation disorders: Fundamental approaches reveal complicated pathways. Am. J. Hum. Genet. 2014, 94, 161–175. [Google Scholar] [CrossRef] [Green Version]
  90. Cantarel, B.L.; Coutinho, P.M.; Rancurel, C.; Bernard, T.; Lombard, V.; Henrissat, B. The Carbohydrate-Active EnZymes database (CAZy): An expert resource for Glycogenomics. Nucleic Acids Res. 2009, 37, D233–D238. [Google Scholar] [CrossRef]
  91. Jaeken, J.; Carchon, H. The carbohydrate-deficient glycoprotein syndromes: An overview. J. Inherit. Metab. Dis. 1993, 16, 813–820. [Google Scholar] [CrossRef]
  92. Rutishauser, U.; Sachs, L. Cell-to-cell binding induced by different lectins. J. Cell Biol. 1975, 65, 247–257. [Google Scholar] [CrossRef]
  93. Audi, J.; Belson, M.; Patel, M.; Schier, J.; Osterloh, J. Ricin poisoning: A comprehensive review. JAMA 2005, 294, 2342–2351. [Google Scholar] [CrossRef]
  94. Schaffrath, R.; Schmitt, M.J.; SpringerLink (Online service). Microbial Protein Toxins. In Topics in Current Genetics; Springer: Berlin/Heidelberg, Germany, 2005; Volume xiii, 237p. [Google Scholar]
  95. Baenziger, J.U.; Fiete, D. Structural determinants of Ricinus communis agglutinin and toxin specificity for oligosaccharides. J. Biol. Chem. 1979, 254, 9795–9799. [Google Scholar] [CrossRef]
  96. Taubenschmid, J.; Stadlmann, J.; Jost, M.; Klokk, T.I.; Rillahan, C.D.; Leibbrandt, A.; Mechtler, K.; Paulson, J.C.; Jude, J.; Zuber, J.; et al. A vital sugar code for ricin toxicity. Cell Res. 2017, 27, 1351–1364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Morgens, D.W.; Wainberg, M.; Boyle, E.A.; Ursu, O.; Araya, C.L.; Tsui, C.K.; Haney, M.S.; Hess, G.T.; Han, K.; Jeng, E.E.; et al. Genome-scale measurement of off-target activity using Cas9 toxicity in high-throughput screens. Nat. Commun. 2017, 8, 15178. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Bassik, M.C.; Kampmann, M.; Lebbink, R.J.; Wang, S.; Hein, M.Y.; Poser, I.; Weibezahn, J.; Horlbeck, M.A.; Chen, S.; Mann, M.; et al. A systematic mammalian genetic interaction map reveals pathways underlying ricin susceptibility. Cell 2013, 152, 909–922. [Google Scholar] [CrossRef] [Green Version]
  99. Moreau, D.; Kumar, P.; Wang, S.C.; Chaumet, A.; Chew, S.Y.; Chevalley, H.; Bard, F. Genome-wide RNAi screens identify genes required for Ricin and PE intoxications. Dev. Cell 2011, 21, 231–244. [Google Scholar] [CrossRef] [Green Version]
  100. Chen, W.; Tang, J.; Stanley, P. Suppressors of alpha(1,3)fucosylation identified by expression cloning in the LEC11B gain-of-function CHO mutant. Glycobiology 2005, 15, 259–269. [Google Scholar] [CrossRef] [Green Version]
  101. Elling, U.; Taubenschmid, J.; Wirnsberger, G.; O’Malley, R.; Demers, S.P.; Vanhaelen, Q.; Shukalyuk, A.I.; Schmauss, G.; Schramek, D.; Schnuetgen, F.; et al. Forward and reverse genetics through derivation of haploid mouse embryonic stem cells. Cell Stem Cell 2011, 9, 563–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Potvin, B.; Stanley, P. Activation of two new alpha(1,3)fucosyltransferase activities in Chinese hamster ovary cells by 5-azacytidine. Cell Regul. 1991, 2, 989–1000. [Google Scholar] [CrossRef]
  103. Voth, D.E.; Ballard, J.D. Clostridium difficile toxins: Mechanism of action and role in disease. Clin. Microbiol. Rev. 2005, 18, 247–263. [Google Scholar] [CrossRef] [Green Version]
  104. Aktories, K.; Schwan, C.; Jank, T. Clostridium difficile Toxin Biology. Annu. Rev. Microbiol. 2017, 71, 281–307. [Google Scholar] [CrossRef]
  105. Kordus, S.L.; Thomas, A.K.; Lacy, D.B. Clostridioides difficile toxins: Mechanisms of action and antitoxin therapeutics. Nat. Rev. Microbiol. 2022, 20, 285–298. [Google Scholar] [CrossRef]
  106. Jank, T.; Aktories, K. Structure and mode of action of clostridial glucosylating toxins: The ABCD model. Trends Microbiol. 2008, 16, 222–229. [Google Scholar] [CrossRef] [PubMed]
  107. Chen, P.; Lam, K.H.; Liu, Z.; Mindlin, F.A.; Chen, B.; Gutierrez, C.B.; Huang, L.; Zhang, Y.; Hamza, T.; Feng, H.; et al. Structure of the full-length Clostridium difficile toxin B. Nat. Struct. Mol. Biol. 2019, 26, 712–719. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, B.; Basak, S.; Chen, P.; Zhang, C.; Perry, K.; Tian, S.; Yu, C.; Dong, M.; Huang, L.; Bowen, M.E.; et al. Structure and conformational dynamics of Clostridioides difficile toxin A. Life Sci. Alliance 2022, 5, e202201383. [Google Scholar] [CrossRef]
  109. Aminzadeh, A.; Larsen, C.E.; Boesen, T.; Jorgensen, R. High-resolution structure of native toxin A from Clostridioides difficile. EMBO Rep. 2022, 23, e53597. [Google Scholar] [CrossRef] [PubMed]
  110. Krivan, H.C.; Clark, G.F.; Smith, D.F.; Wilkins, T.D. Cell surface binding site for Clostridium difficile enterotoxin: Evidence for a glycoconjugate containing the sequence Gal alpha 1-3Gal beta 1-4GlcNAc. Infect. Immun. 1986, 53, 573–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Tucker, K.D.; Wilkins, T.D. Toxin A of Clostridium difficile binds to the human carbohydrate antigens I, X, and Y. Infect. Immun. 1991, 59, 73–78. [Google Scholar] [CrossRef] [Green Version]
  112. Teneberg, S.; Lonnroth, I.; Torres Lopez, J.F.; Galili, U.; Halvarsson, M.O.; Angstrom, J.; Karlsson, K.A. Molecular mimicry in the recognition of glycosphingolipids by Gal alpha 3 Gal beta 4 GlcNAc beta-binding Clostridium difficile toxin A, human natural anti alpha-galactosyl IgG and the monoclonal antibody Gal-13: Characterization of a binding-active human glycosphingolipid, non-identical with the animal receptor. Glycobiology 1996, 6, 599–609. [Google Scholar] [CrossRef] [Green Version]
  113. Tao, L.; Zhang, J.; Meraner, P.; Tovaglieri, A.; Wu, X.; Gerhard, R.; Zhang, X.; Stallcup, W.B.; Miao, J.; He, X.; et al. Frizzled proteins are colonic epithelial receptors for C. difficile toxin B. Nature 2016, 538, 350–355. [Google Scholar] [CrossRef] [Green Version]
  114. Luo, J.; Yang, Q.; Zhang, X.; Zhang, Y.; Wan, L.; Zhan, X.; Zhou, Y.; He, L.; Li, D.; Jin, D.; et al. TFPI is a colonic crypt receptor for TcdB from hypervirulent clade 2 C. difficile. Cell 2022, 185, 980–994.e15. [Google Scholar] [CrossRef]
  115. Lee, H.; Beilhartz, G.L.; Kucharska, I.; Raman, S.; Cui, H.; Lam, M.H.Y.; Liang, H.; Rubinstein, J.L.; Schramek, D.; Julien, J.P.; et al. Recognition of Semaphorin Proteins by P. sordellii Lethal Toxin Reveals Principles of Receptor Specificity in Clostridial Toxins. Cell 2020, 182, 345–356.e16. [Google Scholar] [CrossRef]
  116. Kreuger, J.; Kjellen, L. Heparan sulfate biosynthesis: Regulation and variability. J. Histochem. Cytochem. 2012, 60, 898–907. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Zhou, Y.; Li, D.; Luo, J.; Chen, A.; Li, X.; Pan, Z.; Wan, L.; He, L.; Li, D.; Li, Y.; et al. Sulfated glycosaminoglycans and low-density lipoprotein receptor mediate the cellular entry of Clostridium novyi alpha-toxin. Cell Res. 2021, 31, 935–938. [Google Scholar] [CrossRef]
  118. Iozzo, R.V. Matrix proteoglycans: From molecular design to cellular function. Annu. Rev. Biochem. 1998, 67, 609–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Mizumoto, S.; Yamada, S. Congenital Disorders of Deficiency in Glycosaminoglycan Biosynthesis. Front. Genet. 2021, 12, 717535. [Google Scholar] [CrossRef] [PubMed]
  120. Yuan, P.; Zhang, H.; Cai, C.; Zhu, S.; Zhou, Y.; Yang, X.; He, R.; Li, C.; Guo, S.; Li, S.; et al. Chondroitin sulfate proteoglycan 4 functions as the cellular receptor for Clostridium difficile toxin B. Cell Res. 2015, 25, 157–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Chen, P.; Tao, L.; Wang, T.; Zhang, J.; He, A.; Lam, K.H.; Liu, Z.; He, X.; Perry, K.; Dong, M.; et al. Structural basis for recognition of frizzled proteins by Clostridium difficile toxin B. Science 2018, 360, 664–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Chen, P.; Zeng, J.; Liu, Z.; Thaker, H.; Wang, S.; Tian, S.; Zhang, J.; Tao, L.; Gutierrez, C.B.; Xing, L.; et al. Structural basis for CSPG4 as a receptor for TcdB and a therapeutic target in Clostridioides difficile infection. Nat. Commun. 2021, 12, 3748. [Google Scholar] [CrossRef]
  123. Chen, P.; Jin, R. Receptor binding mechanisms of Clostridioides difficile toxin B and implications for therapeutics development. FEBS J. 2023, 290, 962–969. [Google Scholar] [CrossRef]
  124. Jiang, M.; Shin, J.; Simeon, R.; Chang, J.Y.; Meng, R.; Wang, Y.; Shinde, O.; Li, P.; Chen, Z.; Zhang, J. Structural dynamics of receptor recognition and pH-induced dissociation of full-length Clostridioides difficile Toxin B. PLoS Biol. 2022, 20, e3001589. [Google Scholar] [CrossRef]
  125. Henkel, D.; Tatge, H.; Schottelndreier, D.; Tao, L.; Dong, M.; Gerhard, R. Receptor Binding Domains of TcdB from Clostridioides difficile for Chondroitin Sulfate Proteoglycan-4 and Frizzled Proteins Are Functionally Independent and Additive. Toxins 2020, 12, 736. [Google Scholar] [CrossRef]
  126. Shen, E.; Zhu, K.; Li, D.; Pan, Z.; Luo, Y.; Bian, Q.; He, L.; Song, X.; Zhen, Y.; Jin, D.; et al. Subtyping analysis reveals new variants and accelerated evolution of Clostridioides difficile toxin B. Commun. Biol. 2020, 3, 347. [Google Scholar] [CrossRef]
  127. Mansfield, M.J.; Tremblay, B.J.; Zeng, J.; Wei, X.; Hodgins, H.; Worley, J.; Bry, L.; Dong, M.; Doxey, A.C. Phylogenomics of 8,839 Clostridioides difficile genomes reveals recombination-driven evolution and diversification of toxin A and B. PLoS Pathog. 2020, 16, e1009181. [Google Scholar] [CrossRef] [PubMed]
  128. Wood, J.P.; Ellery, P.E.; Maroney, S.A.; Mast, A.E. Biology of tissue factor pathway inhibitor. Blood 2014, 123, 2934–2943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Mast, A.E. Tissue Factor Pathway Inhibitor: Multiple Anticoagulant Activities for a Single Protein. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 9–14. [Google Scholar] [CrossRef] [Green Version]
  130. Kinoshita, T.; Fujita, M. Biosynthesis of GPI-anchored proteins: Special emphasis on GPI lipid remodeling. J. Lipid Res. 2016, 57, 6–24. [Google Scholar] [CrossRef] [Green Version]
  131. Ferguson, M.A.J.; Hart, G.W.; Kinoshita, T. Glycosylphosphatidylinositol Anchors. In Essentials of Glycobiology, 3rd ed.; Varki, A., Cummings, R.D., Esko, J.D., Stanley, P., Hart, G.W., Aebi, M., Darvill, A.G., Kinoshita, T., Packer, N.H., Prestegard, J.H., et al., Eds.; Cold Spring Harbor (NY): New York, NY, USA, 2015; pp. 137–150. [Google Scholar]
  132. Yamaji, T.; Hanamatsu, H.; Sekizuka, T.; Kuroda, M.; Iwasaki, N.; Ohnishi, M.; Furukawa, J.I.; Yahiro, K.; Hanada, K. A CRISPR Screen Using Subtilase Cytotoxin Identifies SLC39A9 as a Glycan-Regulating Factor. iScience 2019, 15, 407–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Byres, E.; Paton, A.W.; Paton, J.C.; Lofling, J.C.; Smith, D.F.; Wilce, M.C.; Talbot, U.M.; Chong, D.C.; Yu, H.; Huang, S.; et al. Incorporation of a non-human glycan mediates human susceptibility to a bacterial toxin. Nature 2008, 456, 648–652. [Google Scholar] [CrossRef] [Green Version]
  134. Tromp, A.T.; Van Gent, M.; Jansen, J.P.; Scheepmaker, L.M.; Velthuizen, A.; De Haas, C.J.C.; Van Kessel, K.P.M.; Bardoel, B.W.; Boettcher, M.; McManus, M.T.; et al. Host-Receptor Post-Translational Modifications Refine Staphylococcal Leukocidin Cytotoxicity. Toxins 2020, 12, 106. [Google Scholar] [CrossRef] [Green Version]
  135. Spaan, A.N.; Henry, T.; van Rooijen, W.J.M.; Perret, M.; Badiou, C.; Aerts, P.C.; Kemmink, J.; de Haas, C.J.C.; van Kessel, K.P.M.; Vandenesch, F.; et al. The staphylococcal toxin Panton-Valentine Leukocidin targets human C5a receptors. Cell Host Microbe 2013, 13, 584–594. [Google Scholar] [CrossRef] [Green Version]
  136. Spaan, A.N.; Vrieling, M.; Wallet, P.; Badiou, C.; Reyes-Robles, T.; Ohneck, E.A.; Benito, Y.; de Haas, C.J.; Day, C.J.; Jennings, M.P.; et al. The staphylococcal toxins gamma-haemolysin AB and CB differentially target phagocytes by employing specific chemokine receptors. Nat. Commun. 2014, 5, 5438. [Google Scholar] [CrossRef] [Green Version]
  137. Reyes-Robles, T.; Alonzo, F., 3rd; Kozhaya, L.; Lacy, D.B.; Unutmaz, D.; Torres, V.J. Staphylococcus aureus leukotoxin ED targets the chemokine receptors CXCR1 and CXCR2 to kill leukocytes and promote infection. Cell Host Microbe 2013, 14, 453–459. [Google Scholar] [CrossRef] [Green Version]
  138. Drabavicius, G.; Daelemans, D. Intermedilysin cytolytic activity depends on heparan sulfates and membrane composition. PLoS Genet. 2021, 17, e1009387. [Google Scholar] [CrossRef]
  139. Wade, K.R.; Hotze, E.M.; Kuiper, M.J.; Morton, C.J.; Parker, M.W.; Tweten, R.K. An intermolecular electrostatic interaction controls the prepore-to-pore transition in a cholesterol-dependent cytolysin. Proc. Natl. Acad. Sci. USA 2015, 112, 2204–2209. [Google Scholar] [CrossRef]
  140. Lawrence, S.L.; Gorman, M.A.; Feil, S.C.; Mulhern, T.D.; Kuiper, M.J.; Ratner, A.J.; Tweten, R.K.; Morton, C.J.; Parker, M.W. Structural Basis for Receptor Recognition by the Human CD59-Responsive Cholesterol-Dependent Cytolysins. Structure 2016, 24, 1488–1498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Shahi, I.; Llaneras, C.N.; Perelman, S.S.; Torres, V.J.; Ratner, A.J. Genome-Wide CRISPR-Cas9 Screen Does Not Identify Host Factors Modulating Streptococcus agalactiae beta-Hemolysin/Cytolysin-Induced Cell Death. Microbiol. Spectr. 2022, 10, e0218621. [Google Scholar] [CrossRef] [PubMed]
  142. Piper, S.J.; Brillault, L.; Rothnagel, R.; Croll, T.I.; Box, J.K.; Chassagnon, I.; Scherer, S.; Goldie, K.N.; Jones, S.A.; Schepers, F.; et al. Cryo-EM structures of the pore-forming A subunit from the Yersinia entomophaga ABC toxin. Nat. Commun. 2019, 10, 1952. [Google Scholar] [CrossRef] [Green Version]
  143. Roderer, D.; Brocker, F.; Sitsel, O.; Kaplonek, P.; Leidreiter, F.; Seeberger, P.H.; Raunser, S. Glycan-dependent cell adhesion mechanism of Tc toxins. Nat. Commun. 2020, 11, 2694. [Google Scholar] [CrossRef]
  144. Song, N.; Chen, L.; Ren, X.; Waterfield, N.R.; Yang, J.; Yang, G. N-Glycans and sulfated glycosaminoglycans contribute to the action of diverse Tc toxins on mammalian cells. PLoS Pathog. 2021, 17, e1009244. [Google Scholar] [CrossRef] [PubMed]
  145. Xu, Y.; Viswanatha, R.; Sitsel, O.; Roderer, D.; Zhao, H.; Ashwood, C.; Voelcker, C.; Tian, S.; Raunser, S.; Perrimon, N.; et al. CRISPR screens in Drosophila cells identify Vsg as a Tc toxin receptor. Nature 2022, 610, 349–355. [Google Scholar] [CrossRef]
  146. Stechmann, B.; Bai, S.K.; Gobbo, E.; Lopez, R.; Merer, G.; Pinchard, S.; Panigai, L.; Tenza, D.; Raposo, G.; Beaumelle, B.; et al. Inhibition of retrograde transport protects mice from lethal ricin challenge. Cell 2010, 141, 231–242. [Google Scholar] [CrossRef]
  147. Yarwood, R.; Hellicar, J.; Woodman, P.G.; Lowe, M. Membrane trafficking in health and disease. Dis. Model. Mech. 2020, 13, dmm043448. [Google Scholar] [CrossRef] [PubMed]
  148. Selyunin, A.S.; Iles, L.R.; Bartholomeusz, G.; Mukhopadhyay, S. Genome-wide siRNA screen identifies UNC50 as a regulator of Shiga toxin 2 trafficking. J. Cell Biol. 2017, 216, 3249–3262. [Google Scholar] [CrossRef] [PubMed]
  149. Virreira Winter, S.; Zychlinsky, A.; Bardoel, B.W. Genome-wide CRISPR screen reveals novel host factors required for Staphylococcus aureus alpha-hemolysin-mediated toxicity. Sci. Rep. 2016, 6, 24242. [Google Scholar] [CrossRef] [Green Version]
  150. Kouzel, I.U.; Kehl, A.; Berger, P.; Liashkovich, I.; Steil, D.; Makalowski, W.; Suzuki, Y.; Pohlentz, G.; Karch, H.; Mellmann, A.; et al. RAB5A and TRAPPC6B are novel targets for Shiga toxin 2a inactivation in kidney epithelial cells. Sci. Rep. 2020, 10, 4945. [Google Scholar] [CrossRef] [Green Version]
  151. Michalska, M.; Wolf, P. Pseudomonas Exotoxin A: Optimized by evolution for effective killing. Front. Microbiol. 2015, 6, 963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Tafesse, F.G.; Guimaraes, C.P.; Maruyama, T.; Carette, J.E.; Lory, S.; Brummelkamp, T.R.; Ploegh, H.L. GPR107, a G-protein-coupled receptor essential for intoxication by Pseudomonas aeruginosa exotoxin A, localizes to the Golgi and is cleaved by furin. J. Biol. Chem. 2014, 289, 24005–24018. [Google Scholar] [CrossRef] [Green Version]
  153. Chang, S.J.; Jin, S.C.; Jiao, X.; Galan, J.E. Unique features in the intracellular transport of typhoid toxin revealed by a genome-wide screen. PLoS Pathog. 2019, 15, e1007704. [Google Scholar] [CrossRef] [Green Version]
  154. Tian, S.; Wu, Q.; Zhou, B.; Choi, M.Y.; Ding, B.; Yang, W.; Dong, M. Proteomic Analysis Identifies Membrane Proteins Dependent on the ER Membrane Protein Complex. Cell Rep. 2019, 28, 2517–2526.e2515. [Google Scholar] [CrossRef] [PubMed]
  155. Anwar, M.U.; Sergeeva, O.A.; Abrami, L.; Mesquita, F.S.; Lukonin, I.; Amen, T.; Chuat, A.; Capolupo, L.; Liberali, P.; D’Angelo, G.; et al. ER-Golgi-localized proteins TMED2 and TMED10 control the formation of plasma membrane lipid nanodomains. Dev. Cell 2022, 57, 2334–2346.e2338. [Google Scholar] [CrossRef] [PubMed]
  156. Jeon, Y.; Chow, S.H.; Stuart, I.; Weir, A.; Yeung, A.T.; Hale, C.; Sridhar, S.; Dougan, G.; Vince, J.E.; Naderer, T. FBXO11 governs macrophage cell death and inflammation in response to bacterial toxins. Life Sci. Alliance 2023, 6, e202201735. [Google Scholar] [CrossRef]
  157. Alouf, J.E. Bacterial protein toxins. An overview. Methods Mol. Biol. 2000, 145, 1–26. [Google Scholar] [CrossRef] [PubMed]
  158. Gill, D.M. Bacterial toxins: A table of lethal amounts. Microbiol. Rev. 1982, 46, 86–94. [Google Scholar] [CrossRef] [PubMed]
  159. Tian, S.; Liu, Y.; Appleton, E.; Wang, H.; Church, G.M.; Dong, M. Targeted intracellular delivery of Cas13 and Cas9 nucleases using bacterial toxin-based platforms. Cell Rep. 2022, 38, 110476. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The mechanism of action of representative toxins. Pore-forming toxins directly act on the plasma membrane. In contrast, retrograde trafficking toxins and single-chain toxins need to enter the cell through a series of processes, including receptor binding, endocytosis, trafficking/translocation, release/activation, and eventually acting on their cytoplasmic substrates. This process requires multiple host factors, such as receptors, trafficking factors, and translocation factors. ER, endoplasmic reticulum; Stx, Shiga toxin; Ctx, cholera toxin; Ptx, pertussis toxin; EtA, Pseudomonas aeruginosa exotoxin A; SubAB, Escherichia coli subtilase cytotoxin; Cdt, cytolethal distending toxin; BoNT, botulinum neurotoxin; TeNT, tetanus neurotoxin; DT, diphtheria toxin; LCTs, large clostridial toxins; TcdA, Clostridioides difficile toxin A; TcdB, Clostridioides difficile toxin B; TcnA, Clostridium novyi alpha-toxin; CDCs, cholesterol-dependent cytolysins; ILY, Streptococcus intermedius intermedilysin; αHL, Staphylococcus aureus α-hemolysin; Epxs, Enterococcus pore-forming toxins; PVL, Staphylococcus aureus Panton-Valentine leucocidin; HlgCB, Staphylococcus aureus γ-haemolysin CB.
Figure 1. The mechanism of action of representative toxins. Pore-forming toxins directly act on the plasma membrane. In contrast, retrograde trafficking toxins and single-chain toxins need to enter the cell through a series of processes, including receptor binding, endocytosis, trafficking/translocation, release/activation, and eventually acting on their cytoplasmic substrates. This process requires multiple host factors, such as receptors, trafficking factors, and translocation factors. ER, endoplasmic reticulum; Stx, Shiga toxin; Ctx, cholera toxin; Ptx, pertussis toxin; EtA, Pseudomonas aeruginosa exotoxin A; SubAB, Escherichia coli subtilase cytotoxin; Cdt, cytolethal distending toxin; BoNT, botulinum neurotoxin; TeNT, tetanus neurotoxin; DT, diphtheria toxin; LCTs, large clostridial toxins; TcdA, Clostridioides difficile toxin A; TcdB, Clostridioides difficile toxin B; TcnA, Clostridium novyi alpha-toxin; CDCs, cholesterol-dependent cytolysins; ILY, Streptococcus intermedius intermedilysin; αHL, Staphylococcus aureus α-hemolysin; Epxs, Enterococcus pore-forming toxins; PVL, Staphylococcus aureus Panton-Valentine leucocidin; HlgCB, Staphylococcus aureus γ-haemolysin CB.
Bioengineering 10 00884 g001
Figure 2. Forward genetic screen. (a) The screens can be conducted either in a loss-of-function or gain-of-function manner. (b) Schematic diagram of a representative genome-wide CRISPR-Cas9-mediated loss-of-function screen for a toxin.
Figure 2. Forward genetic screen. (a) The screens can be conducted either in a loss-of-function or gain-of-function manner. (b) Schematic diagram of a representative genome-wide CRISPR-Cas9-mediated loss-of-function screen for a toxin.
Bioengineering 10 00884 g002
Figure 3. Scheme of GSL’s biosynthesis pathway and related factors. The abbreviations of GSLs recommended by IUPAC [40] are used in this figure. It highlights the Stxs receptor Gb3, Ctx receptor GM1, and newly identified factors through recent CRISPR screens. GalNAc, N-acetylgalactosamine; GlcNAc, N-acetylglucosamine.
Figure 3. Scheme of GSL’s biosynthesis pathway and related factors. The abbreviations of GSLs recommended by IUPAC [40] are used in this figure. It highlights the Stxs receptor Gb3, Ctx receptor GM1, and newly identified factors through recent CRISPR screens. GalNAc, N-acetylgalactosamine; GlcNAc, N-acetylglucosamine.
Bioengineering 10 00884 g003
Figure 4. Scheme of the major types of protein glycosylation. (a) N-linked glycoprotein. ALGs (asparagine-linked glycosylation enzymes) are required for producing the dolichol-linked precursor oligosaccharide; OSTs (oligosaccharyltransferase complex subunits) are required for transferring the precursor oligosaccharide to an asparagine residue (marked as N) of a protein; a series of glycosidases and glycosyltransferases (e.g., MAN1A2/2A1 and MGAT1/2) are required for converting the precursor oligosaccharide to high-mannose-type or complex-type glycans. (b) O-linked glycoprotein. Four dominant core structures (Core 1to 4) are linked to a serine or threonine residue (marked as S or T) of a protein by a series of glycosyltransferases (e.g., C3GNT and C1GALT1). (c) Proteoglycan. Three representative sGAGs are linked to a serine residue (marked as S) of a core protein by a series of glycosyltransferases and sulfotransferases (e.g., EXT2/3, EXTL1/3, and NDST1/2). (d) GPI-anchored protein. PIGs (phosphatidylinositol glycan enzymes) are required for producing and ligating a GPI anchor to the C-terminus of a protein. Carbohydrate legends are shown at the bottom. GlcN, glucosamine; GlcA, glucuronic acid, IdoA, iduronic acid.
Figure 4. Scheme of the major types of protein glycosylation. (a) N-linked glycoprotein. ALGs (asparagine-linked glycosylation enzymes) are required for producing the dolichol-linked precursor oligosaccharide; OSTs (oligosaccharyltransferase complex subunits) are required for transferring the precursor oligosaccharide to an asparagine residue (marked as N) of a protein; a series of glycosidases and glycosyltransferases (e.g., MAN1A2/2A1 and MGAT1/2) are required for converting the precursor oligosaccharide to high-mannose-type or complex-type glycans. (b) O-linked glycoprotein. Four dominant core structures (Core 1to 4) are linked to a serine or threonine residue (marked as S or T) of a protein by a series of glycosyltransferases (e.g., C3GNT and C1GALT1). (c) Proteoglycan. Three representative sGAGs are linked to a serine residue (marked as S) of a core protein by a series of glycosyltransferases and sulfotransferases (e.g., EXT2/3, EXTL1/3, and NDST1/2). (d) GPI-anchored protein. PIGs (phosphatidylinositol glycan enzymes) are required for producing and ligating a GPI anchor to the C-terminus of a protein. Carbohydrate legends are shown at the bottom. GlcN, glucosamine; GlcA, glucuronic acid, IdoA, iduronic acid.
Bioengineering 10 00884 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Tian, S.; Zhou, N. Gaining New Insights into Fundamental Biological Pathways by Bacterial Toxin-Based Genetic Screens. Bioengineering 2023, 10, 884. https://doi.org/10.3390/bioengineering10080884

AMA Style

Tian S, Zhou N. Gaining New Insights into Fundamental Biological Pathways by Bacterial Toxin-Based Genetic Screens. Bioengineering. 2023; 10(8):884. https://doi.org/10.3390/bioengineering10080884

Chicago/Turabian Style

Tian, Songhai, and Nini Zhou. 2023. "Gaining New Insights into Fundamental Biological Pathways by Bacterial Toxin-Based Genetic Screens" Bioengineering 10, no. 8: 884. https://doi.org/10.3390/bioengineering10080884

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop