Fact-checked by Grok 2 weeks ago

Type III secretion system

The Type III secretion system (T3SS) is a sophisticated nanomachine employed by many to deliver effector proteins directly into the of eukaryotic host cells, enabling precise manipulation of host cellular processes such as immune responses, cytoskeletal dynamics, and to facilitate bacterial survival, replication, or persistence. First discovered in the early in pathogens like . This syringe-like apparatus, also known as an injectisome, spans the bacterial inner and outer membranes, protrudes through a needle filament, and forms a translocation pore in the host , allowing the translocation of partially unfolded effectors without their exposure to the extracellular environment. T3SSs are evolutionarily related to bacterial flagellar systems, sharing conserved core components, and are encoded by gene clusters often acquired via horizontal transfer, resulting in diverse subtypes adapted to specific host interactions. Structurally, the T3SS comprises over 20 proteins organized into a multi-megadalton , including a with concentric rings (such as the inner ring formed by SctD and SctJ, and the outer SctC), an export apparatus driven by an (SctN), a sorting platform (C-ring with SctQ), a hollow needle (polymerized SctF protein, typically 48–80 long), a tip (SctI), and a translocon in the composed of SctB and SctE proteins. Assembly occurs in a hierarchical manner, beginning with the inner membrane export machinery and progressing outward, powered by and the proton motive force, with specialized chaperones maintaining substrate proteins in a secretion-competent, unfolded state. is tightly controlled to ensure temporal specificity: early substrates build the needle, while a "switch" to late substrates (effectors) is triggered by host cell contact, environmental cues like low oxygen or salts, and molecular rulers that measure needle length to prevent overextension. In bacterial , T3SSs are critical factors in numerous human, animal, and plant pathogens, including Yersinia pestis (plague), Salmonella enterica (typhoid fever), Shigella flexneri (dysentery), and Pseudomonas aeruginosa (opportunistic infections), where effectors disrupt , induce , or promote intracellular survival. Beyond disease, T3SSs play essential roles in symbiotic relationships, such as in rhizobial bacteria (Rhizobium spp.) that nodulate plant roots for , highlighting their versatility in trans-kingdom signaling. Due to their indispensability for infection without contributing to essential bacterial physiology, T3SSs represent promising targets for antivirulence therapeutics, vaccines, and engineered protein delivery tools in .

Introduction

Definition and overview

The Type III secretion system (T3SS) is a specialized nanomachine found in , functioning as a molecular that spans the inner and outer bacterial membranes to deliver effector proteins directly from the bacterial into the of eukaryotic host cells. This contact-dependent apparatus, often termed an injectisome, enables to manipulate host cellular processes with high precision, bypassing extracellular exposure that could trigger immune responses. The basic function of the T3SS involves hierarchical protein export, where substrates are secreted in a regulated sequence: first the structural components that assemble the injectisome, followed by translocator proteins that form a in the host , and finally effector proteins that alter host physiology, such as inhibiting or promoting bacterial invasion. This stepwise process ensures efficient assembly and deployment, powered by and the proton motive force across the bacterial . As a major determinant, the T3SS plays a critical role in infections caused by pathogens like , species, and , facilitating host cell subversion while evading detection by the through direct intracellular delivery. T3SSs are widespread, identified in over 100 bacterial genera across at least five phyla, including both and pathogens, underscoring their evolutionary conservation and broad impact on host-bacteria interactions. Unlike Type I and Type II secretion systems, which export proteins into the extracellular milieu via general or dedicated pathways, the T3SS is uniquely injectisome-based and requires intimate bacterial-host contact for translocation, distinguishing it from Type IV (conjugative), Type V (autotransporter), and Type VI (contractile) systems that employ different mechanisms for effector release or delivery.

Discovery and historical context

The initial discovery of the Type III secretion system (T3SS) emerged from genetic studies on bacterial invasion mechanisms in the early 1990s. In , the chromosomal inv locus was identified in 1990 as a key determinant of epithelial cell invasion, with and sequencing revealing the encoding invasin, a surface protein promoting bacterial uptake into host cells. Similarly, in typhimurium, the inv and spa loci, located on Salmonella pathogenicity island 1 (SPI-1), were characterized in 1991 as essential for promoting bacterial entry into cultured intestinal cells, marking early recognition of these loci as invasion factors. These findings built on vague observations from the 1960s of contact-dependent by bacterial pathogens, where direct cell-to-cell contact was noted to trigger host cell damage, though the underlying molecular machinery remained undefined. A pivotal milestone came in 1993 when Jorge E. Galán's laboratory described the T3SS as a specialized "injection machine" capable of directly translocating bacterial proteins into eukaryotic host cells, distinguishing it from other secretion pathways and coining the "type III" terminology based on its unique export mechanism. This insight was reinforced in 1998 by electron microscopy studies on the typhimurium T3SS, which visualized a needle-like appendage protruding from the bacterial envelope and highlighted striking structural and sequence homologies to the flagellar export apparatus, suggesting a shared evolutionary origin for these systems. Throughout the and , further electron microscopy studies provided improved resolutions of the needle complex, including helical reconstructions at near-atomic detail in the 2010s, solidifying the syringe-like model of the T3SS. Pioneering contributions from researchers like Jorge E. Galán, who advanced understanding of effector translocation, and B. Brett Finlay, who identified key T3SS effectors in Salmonella and their roles in modulating host signaling, drove early mechanistic insights into virulence. Early nomenclature was plagued by confusion, as T3SS components shared names with flagellar homologs (e.g., overlapping designations for basal body proteins), complicating comparative studies across bacterial species. This led to a 1998 proposal by Hueck for a unified "Sct" (secretion and cellular translocation) nomenclature, standardizing terms like SctN for the ATPase and SctC for the outer membrane secretin to resolve terminological ambiguities. By 2000, these developments had shifted conceptual understanding from nonspecific contact-induced cytotoxicity to a precise molecular syringe model, laying the foundation for subsequent structural and functional analyses.

Structural Components

Overall architecture

The type III secretion system (T3SS), also known as the injectisome, is a sophisticated syringe-like nanomachine that spans the inner and outer membranes of , enabling the direct injection of effector proteins into host cells. This apparatus assembles hierarchically, beginning with a multiring embedded in the bacterial membranes, which anchors the system and forms the foundation for subsequent components. The overall structure integrates transmembrane elements with extracellular projections, creating a continuous conduit from the bacterial to the host , approximately 100-150 nm in total height depending on the . The hierarchical assembly proceeds from the outward: the base consists of an inner ring complex (composed of proteins like SctD and SctJ) that interfaces with the export apparatus, a -driven inner module (including SctRSTUV and the SctN) responsible for energizing ; this is connected to an outer ring formed by the SctC, typically exhibiting 12- to 15-fold . Protruding from the base is the inner rod (SctI), followed by filament (SctF), a helical with an outer of approximately 8 and an inner channel of about 2.5-3 to accommodate unfolded proteins. The needle terminates in a tip complex (e.g., SctA or pentameric structures like SipD), which senses host contact and facilitates assembly of the translocon—a in the host formed by hydrophobic translocator proteins (e.g., SipB and SipC). In terms of , the needle typically measures 40-80 in length across many , allowing precise host contact without excessive extension, though the full injectisome spans from the bacterial through the and to the host interior. This contrasts with the related bacterial , which shares a conserved core including the inner membrane rings, export apparatus, and but diverges at the extracellular tip: the flagellum features a long, flexible filament for , while the T3SS needle is rigid and optimized for effector delivery rather than propulsion. Visualization of the T3SS often relies on electron microscopy techniques, such as cryo-electron microscopy (cryo-EM) and negative-stain EM, which reveal the characteristic syringe-like projection extending from the bacterial surface upon host contact, with resolutions down to 3-7 Å in recent structures. Species variations in architecture are notable, particularly in needle length: animal pathogens like and exhibit shorter needles (around 40-60 nm) suited to close-range injection, whereas plant pathogens and symbionts display longer filaments, such as 100-200 nm in species, adapting to the thicker walls and extracellular matrices.

Key protein complexes

The Type III secretion system (T3SS) is organized into several key protein complexes that form its modular architecture, analogous to a molecular spanning the bacterial membranes. The serves as the membrane-anchoring foundation, consisting of inner and outer membrane rings that provide structural stability and facilitate assembly. In , the includes inner membrane rings (IR1 and IR2) formed by oligomeric assemblies of proteins such as PrgK and PrgH, which create a multi-ring scaffold spanning the layer, while the outer membrane ring is a β-barrel complex like InvG that embeds in the outer membrane. These rings exhibit concentric oligomeric symmetry, with the inner rings displaying 24-fold in high-resolution structures. The export apparatus forms a specialized within the , comprising a cytoplasmic ring and integral membrane components that control substrate access to the secretion channel. This complex includes a hexameric (SctN) that powers export and is linked to inner membrane proteins forming a selective pore, such as the nonameric export in composed of CdsV in association with the stalk protein CdsO. Structural studies reveal the export apparatus as an extended helical bundle of antiparallel helices from core proteins like SctRST (FliPQR homologs), organizing a central chamber for substrate docking without direct ATP involvement in gating. The needle and tip complex extends extracellularly from the basal body, forming a polymerized for penetration capped by a specialized assembly. The needle is a hollow, helical tube polymerized from proteins like PrgI in , approximately 80 nm long and 7 nm wide, connected to the via an inner rod structure. At the distal end, the tip complex is a pentameric cap formed by translocator proteins such as SipD, which stabilizes the needle and initiates contact, as resolved in cryo-EM structures showing dynamic conformational shifts upon substrate binding. The translocon is a pore-forming that inserts into the host , enabling effector passage, and is composed of amphipathic proteins that oligomerize into a β-barrel-like channel. In , the translocon includes YopB and YopD, which form a heterodimeric with a that disrupts the host , creating a 20-25 as determined by biophysical assays and modeling. This associates with the needle tip via accessory proteins, maintaining a stable conduit across the host . The sorting platform is a cytoplasmic wheel-like scaffold that organizes the hierarchy of substrate export, linking the ATPase to the export apparatus through pod-like substructures. Composed primarily of approximately 24 copies of SctQ (FliM homolog), organized into six pods each with four SctQ subunits, SctL, and SctK, the platform exhibits a cage-like architecture with peripheral pods that dock chaperoned s, as visualized in near-atomic resolution models from . This complex dynamically associates with the inner membrane, facilitating ordered assembly of early, middle, and late s. Recent structural advances, particularly cryo-EM studies achieving resolutions around 3.5-4 , have illuminated dynamic conformational changes across these complexes, such as gating mechanisms in the export apparatus and needle-tip interactions in the SPI-1 T3SS. These high-resolution snapshots, including integrative models of the full needle complex, reveal inter-complex interfaces and flexibility essential for assembly and function.

Protein Components

Classification and nomenclature

The Type III secretion systems (T3SS) are classified into several major families based on phylogenetic analysis of their conserved core proteins, reflecting differences in host specificity and function. These include the Inv/Mxi/Prg family (SPI-1-like systems in enteropathogenic bacteria such as Shigella and Salmonella enterica, involved in intestinal invasion), the Ssa/Esc family (SPI-2-like systems in systemic pathogens like Salmonella and enterohemorrhagic Escherichia coli), the Hrp/Hrc family (prevalent in plant pathogens such as Pseudomonas syringae and Ralstonia solanacearum), the Ysc family (Yersinia-like systems in animal pathogens including Yersinia enterocolitica and Pseudomonas aeruginosa), and the flagellar Fli family (a motility-related system in bacteria like E. coli and Salmonella). Early nomenclature for T3SS components was genus- or species-specific, leading to significant confusion; for example, the ATPase component was named InvA in Salmonella, SpaL in Shigella, and YscN in Yersinia, despite functional homology. This issue was addressed by a unified nomenclature proposed in 1998, which uses the prefix "Sct" (for secretion and cellular translocation) followed by a letter designating the protein class, such as SctC for the outer membrane secretin ring or SctV for the export gate. Orthologs across families are thus denoted consistently (e.g., InvA, SpaS, and YscV all correspond to SctV), facilitating comparative studies and resolving phylogenetic clustering ambiguities. Each T3SS gene cluster typically encompasses 30-50 genes encoding the structural apparatus, export machinery, chaperones, regulators, and associated effectors, often organized as pathogenicity islands. Some bacterial genomes harbor multiple distinct T3SS clusters, enabling adaptation to diverse environments or hosts; for instance, Vibrio parahaemolyticus encodes two separate systems on different chromosomes. Non-canonical T3SS variants also exist, such as versatile symbiotic systems in (e.g., species), which promote nodulation by delivering nodulation-related effectors without pathogenic effects.

Major proteins and their roles

The type III secretion system (T3SS) employs a suite of conserved proteins, standardized under the Sct nomenclature (for secretion and cellular translocation), to orchestrate the assembly of its injectisome and the hierarchical export of substrates. These proteins form distinct functional classes that ensure precise control over protein unfolding, translocation through narrow channels, and substrate specificity during secretion. The SctN is a cytosolic that powers the unfolding and export of substrates by hydrolyzing ATP. It assembles into a hexameric ring structure with six catalytic sites in varying conformational states, facilitating generation and chaperone dissociation from effectors. This energy provision is essential for propelling unfolded proteins through the ~10-Å diameter export channel at rates of up to 60 molecules per second. Chaperones designated as SctL bind to effectors and translocators in the bacterial , preventing their premature aggregation and maintaining them in a secretion-competent, unfolded state. These class I and II chaperones also aid in substrate recognition by the export apparatus and stabilize components of the cytoplasmic sorting platform. By shielding hydrophobic regions, SctL ensures targeted delivery to the for efficient export without off-pathway interactions. The needle protein SctF polymerizes into a , helical filament approximately 50 nm long, serving as a conduit for substrate passage from the bacterial envelope to the host interface. Its assembly occurs via end-on addition of monomers, with length tightly regulated by the secretion rate and molecular rulers to prevent overextension. The needle provides a narrow channel (~20 inner diameter) for substrate transit. The tip protein SctP forms a distal complex that caps the needle, regulates its length as a molecular , and senses host contact, triggering conformational changes to initiate translocon insertion into the eukaryotic . SctP terminates needle upon reaching the appropriate length. The inner protein SctI polymerizes to form the that supports needle . The protein SctW regulates specificity by interacting with the export apparatus, enabling a switch from secretion of early structural components ( and needle) to late effectors upon completion. This control prevents premature effector export and ensures progression. T3SS are secreted in a strict : early such as SctI build the inner ; middle such as SctF assemble the needle; and late include SctP (), translocators, and effectors for host delivery. This temporal ordering is enforced by chaperone interactions and modulation. In species-specific adaptations, such as the SPI-1 T3SS, the protein Spa32 (homologous to SctP) interacts with the export gate to measure and control needle length, coupling polymerization to substrate switching for optimal host penetration.

Secretion Mechanism

Initiation and induction

The initiation of type III secretion systems (T3SS) is triggered by specific environmental cues within the host environment, such as changes in , , and ion concentrations, which signal the bacterium to activate mechanisms. For instance, in , the SPI-2 T3SS is induced by mildly acidic (around 5.5) and low magnesium (Mg²⁺) levels (<2 mM) inside macrophage phagosomes, mediated by the PhoP/PhoQ two-component system that phosphorylates PhoP to upregulate SPI-2 genes including ssrB. Low Mg²⁺ specifically activates PhoQ sensing, leading to enhanced expression of T3SS components essential for intracellular survival. shifts to 37°C, mimicking host conditions, also prime T3SS assembly across many Gram-negative pathogens. As of 2025, studies have further linked AI-2 quorum sensing to regulation of -sensitive PhoP/PhoQ activation in , enhancing acid tolerance and T3SS expression under host conditions. Host cell contact serves as a critical prerequisite for T3SS activation, detected by the needle tip complex, which includes LcrV-like proteins that sense eukaryotic membranes and relieve inhibitory gatekeeper mechanisms. In Yersinia species, the tip complex (e.g., LcrV) undergoes conformational rearrangements upon membrane contact, transmitting a signal through the needle to the basal body and alleviating blockade by gatekeepers like YopN, which otherwise prevents premature effector secretion. This contact-dependent sensing ensures secretion is directed only at target cells, with the tip reorganizing its self-chaperoning domains to switch substrate specificity. Substrate recognition for export begins with chaperone-substrate interactions, where T3SS effectors and translocators bear N-terminal signal peptides of 15-40 unstructured amino acids that direct them to the export gate. These signals, often chaperone-bound (e.g., YscX:YscY heterodimers in Yersinia), engage the export gate (SctV/YscV) via specific motifs, such as C-terminal helices forming salt bridges with gate residues like R701, ensuring ordered delivery while maintaining chaperone-mediated solubility. The export gate's nonameric ring binds these complexes at clefts in its cytosolic domains (SD2-SD4), facilitating proton motive force-dependent translocation. A hierarchical export switch governs the progression from structural component secretion to translocator and effector delivery, completing needle assembly before shifting priorities. In enteropathogenic Escherichia coli, gatekeepers like SepL and SepD initially favor high-affinity binding of translocator-chaperone pairs (e.g., CesAB/EspA, Kd ~0.16 μM) to the translocase EscV, suppressing effector access; their disengagement upon assembly completion lowers the affinity threshold (e.g., to Kd ~0.38 μM for CesT/Tir), enabling effector export. This switch ensures the injectisome matures fully, with early export of rod and needle proteins preceding pore-forming translocators. In Yersinia, calcium-binding regulation further fine-tunes initiation, where low Ca²⁺ concentrations (~0.2 mM) in the host cytosol mimic the low-calcium response, triggering Yop effector secretion by exporting negative regulators like LcrQ and YopD via the T3SS itself. High extracellular Ca²⁺ inhibits this by stabilizing gatekeeper complexes, but depletion leads to growth arrest and massive T3SS upregulation at 37°C, coupling ion sensing to virulence activation. Recent structural studies have illuminated dynamic tip complex behaviors, with integrative modeling revealing conformational flexibility in the needle-tip interface that supports host sensing and substrate switching.

Translocation and effector delivery

The translocation of effector proteins in the Type III secretion system (T3SS) occurs through a narrow channel within the needle filament, which has an inner diameter of approximately 2–3 nm. To pass through this constriction, effectors must be unfolded, a process facilitated by their mechanical lability and powered by ATP hydrolysis from the basal ATPase (SctN), which dechaperones and threads the proteins from the bacterial cytoplasm outward. This export pathway spans the bacterial inner and outer membranes via the basal body, inner rod, and needle, forming a continuous conduit for substrate transit. Upon reaching the needle tip, effectors are delivered into the host cell through the translocon, a pore complex inserted into the eukaryotic plasma membrane. In Pseudomonas aeruginosa, the translocators PopB and PopD assemble into a hetero-oligomeric pore, spanning the host membrane lipid bilayer to enable passage. The translocon attaches to the needle tip (e.g., via PcrV in P. aeruginosa), completing the injectisome and forming a sealed channel for direct cytosol-to-cytosol transfer without intermediate vesicular trafficking. This mechanism allows rapid injection of effector molecules into a host cell shortly after contact, depending on the pathogen and conditions. T3SS effectors lack classical cleavable signal peptides but feature non-cleavable N-terminal secretion signals and chaperone-binding domains (often in the N-terminal region) recognized by dedicated chaperones, which maintain solubility, prevent premature folding, and direct substrates to the export gate with specificity. Chaperone-effector complexes dock at the ATPase-sorting platform, ensuring hierarchical secretion where translocators are exported first to form the pore. The dynamics of translocation involve dual energy sources: the ATPase drives initial unfolding and export of early substrates, while the proton motive force (PMF) across the bacterial inner membrane propels later effectors through the needle and translocon. Recent structural studies (as of 2024) have refined understanding of PMF coupling in substrate propulsion. Needle length adapts to the host cell barrier thickness via molecular ruler proteins (e.g., SctP), optimizing penetration without overextension. Cryo-EM structures from 2023 of the Shigella flexneri T3SS needle complex reveal intricate channel gating mechanisms, including a P/R gasket (formed by residues like F213, M178, and M179) and an R-plug (SpaR residues 111–122) that occlude the export pore to regulate substrate entry and prevent ion leakage during quiescence.

Biological Roles

In pathogenesis

The Type III secretion system (T3SS) plays a central role in bacterial pathogenesis by enabling the delivery of effector proteins that manipulate host cell processes to promote infection and immune evasion. These effectors target key cellular pathways, such as signal transduction and cytoskeletal dynamics, to subvert host defenses and facilitate bacterial survival and replication within the host. For instance, in Yersinia species, the effector YopH acts as a protein tyrosine phosphatase that dephosphorylates host kinases like focal adhesion kinase (FAK) and p130Cas, thereby inhibiting phagocytosis and downstream inflammatory signaling in immune cells. Similarly, T3SS effectors can induce host cell pyroptosis to modulate immune activation; in Salmonella enterica, effectors delivered via the Salmonella pathogenicity island 1 (SPI-1) T3SS, such as SipB, activate caspase-1 to induce pyroptosis in macrophages, releasing pro-inflammatory cytokines like IL-1β while aiding bacterial dissemination by eliminating key immune cells. Effectors also remodel the host cytoskeleton to enhance invasion, as seen with SipA in Salmonella, which binds actin and promotes polymerization to drive membrane ruffling and bacterial uptake into intestinal epithelial cells during gut invasion. Specific pathogens exemplify T3SS-dependent virulence strategies tailored to their infection niches. In Yersinia pestis, the causative agent of bubonic plague, the T3SS injects Yop effectors into macrophages and neutrophils at the dermal inoculation site, blocking phagocytosis and cytokine production to allow systemic spread via lymph nodes without triggering robust inflammation. For Salmonella enterica serovar Typhimurium, the SPI-1 T3SS facilitates gut invasion by promoting effacement of the epithelial barrier and entry into M cells and enterocytes, enabling traversal to deeper tissues and induction of colitis. In Pseudomonas aeruginosa, a major opportunistic pathogen in lung infections, the T3SS delivers effectors like ExoU, a phospholipase that disrupts epithelial barriers and induces rapid cytotoxicity in alveolar cells, exacerbating acute pneumonia in vulnerable hosts such as those with cystic fibrosis. The pathogenesis process unfolds in distinct stages orchestrated by the T3SS. Initial adhesion of bacteria to host cells via surface structures positions the T3SS needle for contact-dependent activation, followed by rapid injection of translocators and effectors through the host plasma membrane. Effector action then inhibits phagocytosis by disrupting actin dynamics or dephosphorylating uptake signaling pathways, while also modulating cytokine production—such as suppressing pro-inflammatory IL-1β or promoting anti-apoptotic signals—to dampen innate immunity and create a permissive niche for replication. Host cells counter T3SS activity through inflammasome-mediated responses, where nucleotide-binding oligomerization domain-like receptors (NLRs) and NAIP proteins detect T3SS components like the needle protein or translocators, assembling the NAIP/NLRC4 inflammasome to activate caspase-1 and trigger pyroptosis—a lytic form of cell death that releases pro-inflammatory contents and restricts bacterial spread. This detection mechanism is crucial in macrophages, where it promotes clearance of infected cells during early infection stages. Disruption of the T3SS severely impairs virulence, with mutants exhibiting 10- to 1000-fold reductions in bacterial loads and survival in animal models of infection; for example, T3SS-deficient Yersinia pestis strains show dramatically increased LD50 values in mouse bubonic plague models, failing to disseminate beyond the inoculation site. Recent research highlights the nuanced roles of T3SS effectors in modulating host-microbe interactions beyond direct cytotoxicity. A 2025 study demonstrated that accessory effectors of the Salmonella SPI-2 T3SS collectively influence gut microbiota composition and dynamics during chronic infection, promoting dysbiosis that favors bacterial persistence and transmission in mouse models of colitis.

Non-pathogenic functions and applications

In symbiotic interactions, the type III secretion system (T3SS) of rhizobia plays a crucial role in establishing nitrogen-fixing associations with legumes by delivering effector proteins, known as nodulation outer proteins (Nops) or type III effectors (T3Es), directly into host plant cells to suppress immunity and promote root nodule formation. For instance, in Rhizobium and Bradyrhizobium species, T3Es such as NopL and NopP suppress plant immune responses, such as by targeting MAPK signaling, to bypass defenses, enabling intracellular infection and nodule organogenesis essential for mutualistic nitrogen fixation. These effectors facilitate host range specificity, with T3SS mutants often showing reduced nodulation efficiency on certain legumes. Beyond symbiosis, T3SS contributes to non-pathogenic environmental functions in bacteria like Vibrio parahaemolyticus, where it enhances survival and fitness in aquatic ecosystems by enabling cytotoxicity against bacterivorous protists such as Cafeteria roenbergensis. T3SS-2 positive strains maintain stable populations and reduce protist densities by over 85% in microcosms, promoting invasion into plankton communities under varying temperatures (16–30°C) and nutrient conditions, thus supporting environmental persistence without host infection. Engineered T3SS has emerged as a versatile platform for biotechnological protein delivery, leveraging its syringe-like mechanism to inject heterologous cargos into eukaryotic cells for applications in vaccines and gene therapy. Non-pathogenic chassis, such as attenuated Escherichia coli strains, have been optimized for cargo injection, thereby enhancing antigen presentation and immune activation. Recent 2025 reviews highlight the use of E. coli Nissle 1917 with inducible T3SS for safe, tunable delivery of therapeutic proteins, minimizing immunogenicity while enabling oral administration. In industrial contexts, T3SS-based systems serve as targeted drug delivery vehicles, particularly for cancer therapy, where engineered bacteria inject pro-apoptotic effectors like the Noxa mitochondrial domain into tumor cells, inducing selective cytotoxicity without affecting healthy tissues. A 2025 study demonstrated the robustness of Salmonella SPI-1 T3SS under simulated microgravity, maintaining comparable protein export levels to Earth conditions (p > 0.05) despite elevated stress responses, positioning it as a promising for space biology applications in and purification. T3SS offers advantages over vectors, including direct cytosolic translocation without endosomal escape requirements, high specificity via host cell contact, and reduced due to transient bacterial presence (cleared within 10 hours), though challenges persist with size limits around 50–60 kDa for efficient . Recent advances in 2025 include T3SS-engineered for delivery, such as injecting host-defense peptides into infected cells to combat multidrug-resistant pathogens, achieving up to 2–3-fold higher efficacy than free peptides in preclinical models.

Regulation and Evolution

Regulatory mechanisms

The regulation of Type III secretion systems (T3SS) in is multifaceted, involving transcriptional, post-transcriptional, and post-translational controls that integrate environmental signals to coordinate expression and activity with stages. These mechanisms ensure precise timing of T3SS deployment, preventing unnecessary energy expenditure and modulating in response to cues. Transcriptional regulation often relies on two-component systems that sense extracellular conditions. In Salmonella enterica, the PhoP/PhoQ system detects low magnesium ion (Mg²⁺) concentrations, activating PhoP to repress SPI-1 T3SS genes while promoting SPI-2 expression inside host cells, thereby switching secretion systems during intracellular phases. Sigma factors further drive operon activation; for instance, in Pseudomonas aeruginosa, the alternative sigma factor RpoN (σ⁵⁴) initiates transcription of T3SS genes under nitrogen-limiting conditions, with the master regulator ExsA binding promoter regions to coordinate the entire regulon. Post-transcriptional control involves small regulatory RNAs (sRNAs) and chaperone-mediated feedback. Chaperone feedback loops, such as the ExsC-ExsD-ExsE circuit in , link secretion activity to ; ExsC relieves ExsD inhibition of ExsA when needles form, amplifying transcription only during active . Activity regulation includes auto-inhibitory loops via translocated effectors. Environmental integration occurs through and phase variation. In , the LuxS-dependent autoinducer AI-2 represses T3SS genes at high cell densities via the LuxR regulator, favoring formation over in crowded environments. Phase variation enables on/off switching; in some enteropathogens, invertible promoters or slipped-strand mispairing toggle T3SS expression stochastically, adapting to fluctuating host niches. Multi-system coordination involves crosstalk between T3SS variants. In Salmonella enterica, SPI-1 and SPI-2 T3SS expression is temporally segregated, with SPI-1 activation in the gut lumen repressing SPI-2 via shared regulators like HilD and SsrB, ensuring sequential invasion and intracellular survival during infection progression.

Evolutionary origins

The type III secretion system (T3SS) and the bacterial share a common evolutionary ancestor, evidenced by and structural conservation among their core components. Approximately 20 core orthologs are conserved between the two systems, including key export apparatus proteins such as SctR (homologous to ) and SctS (homologous to FliQ), which form the inner pore for protein translocation. These similarities extend to the and components, supporting a model where the T3SS diverged from a flagellar around 2-3 billion years ago, predating the emergence of eukaryotic hosts, though alternative hypotheses suggest independent or T3SS as ancestral in some lineages based on phylogenomic analyses. The evolutionary path posits the as the ancestral system, initially evolved for , with the non-flagellar T3SS (NF-T3SS) arising through duplications, deletions, and recruitments that repurposed the export machinery for protein secretion into host cells. This adaptation for involved the loss of -related elements like the and hook, coupled with the addition of a translocon and needle for effector delivery. (HGT) played a pivotal role in this diversification, with T3SS clusters often residing in pathogenicity islands or plasmids that facilitate , particularly in such as Salmonella and Yersinia. Prior to their co-option for , ancestral T3SS precursors likely served non-pathogenic functions, such as nutrient acquisition in environmental or symbiotic interactions in early microbial communities. Phylogenetic analyses indicate these systems originated in ancient Proteobacteria, with secretins (outer membrane channels) recruited multiple times from type II secretion systems to enable cell targeting. Today, this single ancestral system has given rise to over 10 distinct T3SS classes, reflecting extensive HGT and adaptation across .

Research Methods and Tools

Experimental techniques

Several experimental techniques have been developed to isolate and purify components of the type III secretion system (T3SS), enabling detailed studies of its assembly and function. Needle complexes, which form the extracellular portion of the T3SS, are often isolated using methods, such as cesium chloride (CsCl) gradients, to separate intact complexes from cellular debris and other proteins. is another widely used approach for purifying needle complexes, allowing fractionation based on size and while preserving structural integrity. Affinity purification techniques, employing epitope-tagged components like His-tagged export apparatus proteins, have been applied to isolate basal bodies embedded in the bacterial membrane, facilitating downstream biochemical and structural analyses. Structural biology methods have provided high-resolution insights into T3SS architecture. Cryo-electron microscopy (cryo-EM) has been instrumental in visualizing intact needle complexes and injectisomes, achieving resolutions below 4 Å since 2020, as demonstrated in structures of the T3SS at 3.7 Å resolution. is commonly used to determine atomic structures of subcomplexes, such as the sorting platform or individual ring components, complementing cryo-EM data for non-crystalline assemblies. (NMR) spectroscopy has been employed to probe dynamic aspects of T3SS components, including needle filament flexibility and interactions in membrane environments. Functional assays assess T3SS activity and protein interactions. The β-lactamase reporter assay measures translocation efficiency by fusing a β-lactamase to effector proteins; successful injection into host cells cleaves a fluorescent , allowing quantification via or . Yeast two-hybrid screening identifies protein-protein interactions within the T3SS, such as between apparatus components like Ysc proteins in , by detecting activation in yeast cells. Genetic tools enable systematic dissection of T3SS function. Mutant libraries generated via transposon insertion screens, such as Tn-seq in or , identify essential genes for T3SS assembly and secretion by comparing mutant fitness under inducing conditions. systems in facilitate reconstitution of T3SS from pathogens like , allowing controlled delivery of reporter proteins for functional validation. approaches characterize the T3SS secretome. -based analysis of culture supernatants or host cell lysates identifies secreted effectors, as in quantitative proteomic profiling of the T3SS secretome, which confirmed dozens of effectors and chaperones. These methods often combine of tagged substrates with liquid chromatography-tandem (LC-MS/MS) to discover novel effectors. Microscopy techniques visualize T3SS dynamics in real time. Fluorescence microscopy, using GFP-tagged effectors or apparatus components, tracks injection events during host cell contact, revealing spatiotemporal patterns of secretion. , including single-molecule switching techniques, resolves tip dynamics and individual injectisome positioning on the bacterial surface with nanometer precision. Recent advances include single-molecule () to study export kinetics, providing insights into chaperone-effector unfolding and translocation rates at the single-event level.

Computational prediction tools

Computational tools play a crucial role in identifying and annotating components of the type III secretion system (T3SS), particularly for predicting signal peptides, effectors, and structural features in bacterial genomes where experimental data is limited. These bioinformatics approaches leverage sequence features, , and structural modeling to facilitate genome annotation and discovery of novel T3SS elements. Signal peptide predictors focus on N-terminal motifs that direct proteins to the T3SS. EffectiveT3 is a widely used tool that scores potential type III secreted proteins by integrating composition with secondary structure predictions, emphasizing hydrophobicity and charge distributions in the first 50 . Similarly, T3_MM employs a to classify bacterial type III signals based on composition within the N-terminal 100 residues, achieving effective discrimination between T3SS and non-T3SS proteins through probabilistic sequence modeling. For effector identification, genomic island scanning tools like IslandViewer detect T3SS gene clusters within pathogenicity islands by integrating multiple methods for island prediction, aiding in the contextual identification of potential effectors. Motif-based approaches target C-terminal signals characteristic of T3SS effectors; for instance, tools analyzing bipartite patterns in these regions help classify candidates by recognizing conserved motifs associated with . Structure modeling of T3SS components benefits from advanced predictive methods. has been applied to generate high-confidence models of T3SS proteins, such as translocators and chaperones, by predicting folding based on evolutionary couplings in sequences. , often using flagellar assembly templates due to structural similarities between T3SS and flagella, provides insights into needle complex and export apparatus architectures when sequence identity is sufficient. Key databases support these predictions by curating T3SS-related data. T3DB integrates annotated genes, proteins, and orthologs for T3SS apparatus, chaperones, and effectors across bacterial species, serving as a reference for training and validating predictors. These tools generally exhibit around 80-90% for known effectors in datasets, though performance drops for or divergent systems due to reliance on training data from well-studied pathogens. Recent advancements incorporate , such as in DeepT3 models, to enhance predictions for unpublished genomes by learning complex sequence patterns beyond traditional motifs.

Therapeutic inhibitors and developments

The Type III secretion system (T3SS) has emerged as a promising target for anti-virulence therapies against Gram-negative pathogens, aiming to disrupt effector delivery without killing the , thereby minimizing selective pressure for resistance development. Inhibitors primarily target conserved components like the or needle tip to block secretion and translocation, while and approaches neutralize tip proteins to impair injectisome function. These strategies exploit the T3SS's essential role in pathogenesis, such as in lung infections and . Small-molecule inhibitors represent a major class, with salicylidene acylhydrazides like INP0400 blocking T3SS-mediated effector secretion in pathogens including and by interfering with translocation without affecting bacterial growth. ATPase-targeted compounds, such as those inhibiting YscN in or EscN in enteropathogenic , prevent energy-dependent substrate export, reducing virulence in cellular models. Needle tip disruptors, including phenolic compounds like and trans-cinnamic acid, repress T3SS (e.g., hrpA and hrpL) in and related pathogens, thereby inhibiting assembly and injection efficiency. Antibody-based therapies focus on tip proteins, with monoclonal antibodies against LcrV in directly neutralizing effector translocation and enhancing in mouse models of . Subunit incorporating LcrV, often combined with F1 antigen, elicit protective antibodies that reduce T3SS injection, as demonstrated in recombinant and mRNA-lipid nanoparticle formulations protecting mice from bubonic and . Similarly, anti-PcrV antibodies target the T3SS tip to block in lung epithelial cells. High-throughput screening assays, utilizing reporter fusions for effector secretion or activation, have identified novel inhibitors, while structure-based design leverages cryo-EM structures of injectisomes for targeted binding to or tip sites. Clinical progress includes I/II trials of bispecific antibodies like MEDI3902 (anti-PcrV/Psl) for infections in and ventilator-associated pneumonia, showing tolerability but mixed efficacy in reducing bacterial load. vaccine candidates targeting T3SS components, such as SipD, remain in preclinical stages, with ongoing efforts to improve . Key challenges include achieving specificity to avoid off-target effects on host ATPases or non-pathogenic microbiota, as well as potential resistance through T3SS mutations that alter inhibitor binding without restoring full virulence. Bioavailability and delivery in polymicrobial infections further complicate translation. Recent advances, as highlighted in 2024 and 2025 reviews, include broad-spectrum inhibitors like cyclic peptomers and engineered antibodies, alongside links to metabolic therapies where T3SS inhibition via dietary mitigates intestinal barrier disruption in and non-alcoholic . In 2025, de novo designed protein antagonists have emerged as a class of T3SS inhibitors, rationally targeting injectisome components through computational design pipelines.

References

  1. [1]
  2. [2]
  3. [3]
  4. [4]
  5. [5]
  6. [6]
    Sequence, localization and function of the invasin protein of Yersinia ...
    The inv locus of Yersinia enterocolitica is sufficient to convert a non-invasive Escherichia coli K12 strain into a microorganism that is able to penetrate ...Missing: discovery | Show results with:discovery
  7. [7]
    High-resolution structure of the Shigella type-III secretion needle by ...
    Sep 29, 2014 · The structure of the type-III secretion system needle of Shigella flexneri is determined to a precision of 0.4 Å.
  8. [8]
    bacterial devices for protein delivery into host cells - PubMed - NIH
    A complex protein secretion system termed type III to deliver bacterial effector proteins into host cells that then modulate host cellular functions.
  9. [9]
  10. [10]
    The Structure and Function of Type III Secretion Systems
    Type III secretion systems (T3SSs) afford Gram-negative bacteria an intimate means of altering the biology of their eukaryotic hosts.
  11. [11]
    Assembly and architecture of the type III secretion sorting platform
    Type III secretion systems are bacterial nanomachines specialized in protein delivery into target eukaryotic cells. The structural and functional complexity ...
  12. [12]
    Building a secreting nanomachine: a structural overview of the T3SS
    Apr 1, 2014 · The type III secretion system (T3SS), has evolved to provide a means of transporting proteins from the bacterial cytoplasm across the periplasmic and ...
  13. [13]
    Cryo-EM analysis of the T3S injectisome reveals the structure of the ...
    Sep 21, 2018 · The major structural unit of the injectisome is the basal body, composed of three highly oligomerized concentric rings spanning the bacterial ...
  14. [14]
    “The structure of the Type III secretion system export gate with CdsO ...
    Oct 13, 2020 · This work describes the molecular architecture of two essential components of a T3SS, identifies the interface between the components, and ...
  15. [15]
    Structure of the core of the type III secretion system export apparatus
    Jul 2, 2018 · All three subunits form extended structures, built predominantly from pairs of kinked antiparallel helices, with a common orientation of N and C ...
  16. [16]
    Cryo-EM structure of the Shigella type III needle complex
    Feb 24, 2020 · The T3SS needle complex is a syringe-shaped nanomachine consisting of two membrane-embedded ring systems that sheath a central export apparatus and a hollow ...
  17. [17]
    Cryo-EM structure of the needle filament tip complex of the ... - PNAS
    Oct 27, 2021 · Advances in cryoelectron microscopy (cryo-EM) have allowed the visualization of most components of the T3SS machine at high resolution, both in ...
  18. [18]
    Translocators YopB and YopD from Yersinia enterocolitica Form a ...
    The type III secretion systems are contact-activated secretion systems that allow bacteria to inject effector proteins across eukaryotic cell membranes.
  19. [19]
    The type III secretion system needle, tip, and translocon - Dey - 2019
    Jul 13, 2019 · The structural component of the T3SS is a syringe-like needle apparatus (Figure 1) that is assembled from over 20 different proteins, forming a ...
  20. [20]
    Cytosolic sorting platform complexes shuttle type III secretion system ...
    Jan 3, 2024 · While in the flagellum, FliMN forms a continuous ring structure involved in switching the direction of flagellar rotation, six distinct pod ...
  21. [21]
    Structure and Function of the Type III Secretion System of ... - NIH
    The basal body brings effector toxins from the bacterial cytosol to the needle-like structure. It consists of an inner membrane component and an outer membrane ...
  22. [22]
    Phylogenetic analysis of a gene cluster encoding an additional ...
    Sep 2, 2012 · The central role of Type III secretion systems (T3SS) in bacteria-plant interactions is well established, yet unexpected findings are being ...
  23. [23]
    Escherichia coli type III secretion system 2: a new kind of T3SS? - PMC
    SPI-2 T3SS could translocate both effectors encoded in SPI-1 and SPI-2. The ETT2 gene cluster can also regulate genes encoded elsewhere on the chromosome.
  24. [24]
    The Versatile Roles of Type III Secretion Systems in Rhizobium ...
    Sep 8, 2022 · To suppress plant immunity and promote the intracellular infection required for fixing nitrogen for the benefit of their legume hosts, ...
  25. [25]
  26. [26]
  27. [27]
    Cryo-EM structure of the homohexameric T3SS ATPase-central stalk ...
    Feb 7, 2019 · The EscN-EscO structure provides significant atomic insights into how the ATPase contributes to type III secretion, including torque generation ...Missing: SctN | Show results with:SctN
  28. [28]
    The Bacterial Type III Secretion System as a Broadly Applied Protein ...
    Jan 3, 2025 · This review explores recent advancements in the structure and function of the T3SS. We explore the molecular underpinnings of the T3SS apparatus ...
  29. [29]
    The type III secretion system needle tip complex mediates host cell ...
    As the needle is implicated in host cell sensing and secretion regulation, its tip should contain components that initiate host cell contact. Through ...Missing: SctI PQ
  30. [30]
    Bacterial type III secretion systems: a complex device for the delivery ...
    They were coined ... et al. Pseudomonas syringae lytic transglycosylases coregulated with the type III secretion system contribute to the translocation of ...Missing: 1993 | Show results with:1993
  31. [31]
    [PDF] Type III Secretion: Building and Operating a Remarkable ...
    The Type III secretion system (T3SS) is a protein export pathway that is wide- spread in Gram-negative bacteria and delivers effector proteins directly into.<|control11|><|separator|>
  32. [32]
    Bacterial type III secretion systems: a complex device for the delivery ...
    Virulence-associated type III secretion systems (T3SS) serve the injection of bacterial effector proteins into eukaryotic host cells.
  33. [33]
    Timing is everything: the regulation of type III secretion - PMC
    There is little dispute that the Spa32 family of proteins is responsible for needle-length control and substrate switching. ... In Salmonella, it has been shown ...
  34. [34]
    Molecular ruler determines needle length for the Salmonella Spi-1 ...
    Mar 16, 2015 · We present evidence that the Spi-1 injectisome, like the Salmonella flagellar hook, uses a secreted molecular ruler, InvJ, to determine needle length.
  35. [35]
    How the PhoP/PhoQ System Controls Virulence and Mg2+ ...
    Jun 30, 2021 · The PhoP/PhoQ system responds to changes in the Mg2+ concentration in its surroundings by altering expression of different classes of genes ...
  36. [36]
    Assembly and structure of the T3SS - ScienceDirect.com
    Electron microscopy was also used to generate an ~ 18 Å model of the Salmonella typhimurium needle with an average symmetry of 6.3 subunits per turn and a 26 Å ...
  37. [37]
    Direct interaction of a chaperone-bound type III secretion substrate ...
    Jun 2, 2022 · The export gate SctV (YscV in Yersinia) binds substrate:chaperone complexes such as YscX:YscY, which are essential for formation of a functional T3SS.Missing: aa | Show results with:aa
  38. [38]
    Type III secretion systems: the bacterial flagellum and the injectisome
    Oct 5, 2015 · The flagellum and the injectisome are two of the most complex and fascinating bacterial nanomachines. At their core, they share a type III secretion system (T3 ...
  39. [39]
    Hierarchical protein targeting and secretion is controlled by an ...
    Type III secretion (T3S), a protein export pathway common to Gram‐negative pathogens, comprises a trans‐envelope syringe, the injectisome, ...
  40. [40]
    Induction of the Yersinia Type 3 Secretion System as an All-or-None ...
    When extracellular Ca2+ levels fall to the low millimolar range, cytosolic LcrQ and YopD are exported from the bacterial cell by the T3SS, further elevating the ...
  41. [41]
    Integrative structural analysis of the type III secretion system needle ...
    Apr 1, 2023 · We combined cryo‐electron microscopy, cross‐linking mass spectrometry, and integrative modeling to determine the structure of the T3SS needle complex from ...Missing: LcrV changes 2020-2025
  42. [42]
    The Salmonella Type III Secretion System Virulence Effector Forms ...
    Jan 21, 2015 · The diameter of the needle's central channel is ∼2 to 3 nm, which is too small to allow secretion of fully folded effector proteins. A ...
  43. [43]
    Type III secretion system effector proteins are mechanically labile
    Mar 15, 2021 · Type III secretion systems (T3SS) are large nanomachines utilized by both pathogenic and symbiotic bacteria to inject effector proteins directly ...
  44. [44]
    The Structure and Function of Type III Secretion Systems - PMC
    Mar 23, 2016 · Type III secretion systems (T3SS) afford gram-negative bacteria a most intimate means of altering the biology of their eukaryotic hosts ...
  45. [45]
    Frontiers | Protein Secretion Systems in Pseudomonas aeruginosa
    Jul 17, 2011 · The type III secretion system. The T3SS is a fascinating secretion system. In Yersinia enterocolitica, it was proposed to secrete proteins ...
  46. [46]
    PopB-PcrV Interactions Are Essential for Pore Formation ... - PubMed
    Oct 26, 2022 · In Pseudomonas aeruginosa, the translocation pore is formed by proteins PopB and PopD and attaches to the T3SS needle via the needle tip protein PcrV.Missing: beta- barrel
  47. [47]
    Real-Time Analysis of Effector Translocation by the Type III ...
    Feb 14, 2008 · Bacteria use type III secretion systems (TTSS) to translocate effector proteins into host cells. Better understanding of the TTSS and its ...
  48. [48]
    Structural Insights into Type III Secretion Systems of the Bacterial ...
    Sep 15, 2023 · Annual Review of Microbiology Volume 77, 2023 Structural Insights into Type III Secretion Systems of the Bacterial Flagellum and Injectisome
  49. [49]
  50. [50]
    Yersinia type III effectors perturb host innate immune responses - PMC
    To date, it has been reported that YopH binds to and dephosphorylates the focal adhesion kinase (FAK), Crk associated tyrosine kinase substrate (p130Cas), and ...
  51. [51]
    Programmed cell death and Salmonella pathogenesis - NIH
    Jan 4, 2024 · The activation and inhibition of apoptosis, pyroptosis, necroptosis, and canonical autophagy are mediated by the SPI-1 and SPI-2 T3SS effector ...
  52. [52]
    Impact of Salmonella enterica Type III Secretion System Effectors on ...
    Dec 31, 2012 · This paper reviews our current knowledge about the functions, biochemical activities, host targets, and impact on host cells of these effectors.
  53. [53]
    Gain-of-Function Analysis Reveals Important Virulence Roles for the ...
    The Y. pestis type III secretion system (T3SS) is a major contributor to this innate immune evasion strategy. The T3SS transports bacterial proteins, called ...
  54. [54]
    Salmonella effector driven invasion of the gut epithelium
    Sep 6, 2021 · In this review, we summarize recent findings explaining IEC invasion by type-three-secretion-system-1 (TTSS-1) effector proteins and discuss their effects on ...
  55. [55]
    The type III secretion system facilitates systemic infections of ...
    Dec 13, 2023 · Here, we found that the type III secretion system effector ExoU was the main determinant of pathogenicity of a highly virulent clinical isolate strain.
  56. [56]
    Type III Secretion Systems and Disease - PMC - PubMed Central
    This bacterial type III secretion system (T3SS) is of special interest to those studying host-pathogen interactions because, by utilizing this system, bacteria ...Missing: coined 1993
  57. [57]
    Human NAIP/NLRC4 and NLRP3 inflammasomes detect ...
    The NAIP/NLRC4 inflammasome promotes control of intestinal Salmonella infection in mice by triggering pyroptosis and expulsion of infected intestinal epithelial ...
  58. [58]
    The accessory type III secretion system effectors collectively shape ...
    Injection of effectors via a type III secretion system (T3SS) is an infection strategy shared by various Gram-negative bacterial pathogens, many infecting ...<|control11|><|separator|>
  59. [59]
    Hijacking of leguminous nodulation signaling by the rhizobial type III ...
    Sep 30, 2013 · Here we show that symbiosis in the soybean rhizobium Bradyrhizobium elkanii is promoted by the type III secretion system (T3SS), which delivers virulence ...
  60. [60]
    The rhizobial type III effectors ErnA and Sup3 hijack the ...
    Jun 22, 2025 · Flavonoids released by legume roots induce the production of NFs and activate the type III secretion system (T3SS) in rhizobia via NodD and TtsI ...
  61. [61]
    Acquired Type III Secretion System Determines Environmental ...
    Here, we test for the contribution of type III secretion systems (T3SS) to the environmental fitness of epidemic Vibrio parahaemolyticus.
  62. [62]
    Bacterial type III secretion system as a protein delivery tool for a ...
    A protein delivery tool based on bacterial type III secretion system (T3SS) has been broadly applied in biomedical researches.
  63. [63]
    Beyond pathogenicity: applications of the type III secretion system ...
    This comprehensive review analyzes the molecular structure and regulatory mechanisms of T3SS, while also exploring its emerging biomedical ...
  64. [64]
    Engineering Escherichia coli Nissle as safe chassis for delivery of ...
    Aug 30, 2025 · Developing novel tools to engineer safe, nonpathogenic microbial platforms is essential for advancing clinical therapies. In this work, we ...
  65. [65]
    Targeted Delivery of the Mitochondrial Target Domain of Noxa to ...
    Jul 16, 2020 · Here, we tested the potential of synthetic T3SS as drug delivery machinery for peptide-based drugs owing to its modular nature. First, the ...Missing: vehicles | Show results with:vehicles
  66. [66]
    Evaluation of the Salmonella type 3 secretion system (T3SS) as part ...
    Apr 1, 2025 · In this study, we sought to investigate the effects of simulated microgravity on the SPI-1 T3SS as a potential protein production platform for ...
  67. [67]
    LITESEC-T3SS - Light-controlled protein delivery into eukaryotic ...
    May 13, 2020 · However, most cargoes, including large proteins with molecular weights above 60 kDa, can be exported by the T3SS. Protein translocation into ...
  68. [68]
    Bacterial Systems as a Precision Delivery Platform of Therapeutic ...
    Feb 5, 2025 · Once localized, they are capable of releasing therapeutic peptides directly into the tumor microenvironment or even into cancer cells, thereby ...
  69. [69]
    The PhoP/PhoQ system controls the intramacrophage type three ...
    Here we determine that the PhoP/PhoQ two-component system controls the intramacrophage expression of spi/ssa genes by regulating the SsrB/SpiR system.Missing: III | Show results with:III
  70. [70]
    Transcriptional regulation of the Pseudomonas aeruginosa type III ...
    Type III secretion systems (T3SS) function by translocating effector proteins into eukaryotic host cells and are important for the virulence of many ...
  71. [71]
    Global RNA interactome of Salmonella discovers a 5′ UTR sponge ...
    Feb 3, 2022 · Particularly, MicA, CpxQ, MgrR, CyaR, and RybB represent a core of sRNAs involved in the regulation of mRNAs encoding OM or IM proteins (Figure ...
  72. [72]
    Transcriptional Induction of the Pseudomonas aeruginosa Type III ...
    The first mechanism is dependent upon ExsC, a regulatory protein that couples transcription of the T3SS to the activity of the type III secretion machinery.
  73. [73]
    The Salmonella Effector SspH2 Facilitates Spatially Selective ...
    Aug 27, 2024 · We show that SspH2 modulates host innate immunity by interacting with both NOD1 and NOD2 in mammalian epithelial cell culture via the NF-κB ...
  74. [74]
    Quorum Sensing Regulates Type III Secretion in Vibrio harveyi ... - NIH
    We find that at high cell density (in the presence of autoinducers), quorum sensing represses TTS in V. harveyi and V. parahaemolyticus.Missing: utilization modulation
  75. [75]
    Regulation of type III secretion system in Pseudomonas syringae
    Overall, these studies have suggested the existence of a highly intricate regulatory network for T3SS, which controls the pathogenicity of P. syringae. This ...
  76. [76]
    Interrelationship between type three secretion system and ...
    Salmonella species causing typhoid fever and gastroenteritis deploy two T3SSs encoded in Salmonella pathogenicity islands SPI-1 and SPI-2, here termed T3SS-1 ...
  77. [77]
    Type III Secretion System in Intestinal Pathogens and Metabolic ...
    Nov 6, 2024 · This article reviews the structure and function of the T3SS, the disruption of intestinal barrier integrity by the T3SS, the changes in intestinal flora ...3. The T3ss Disrupts... · 4.1. T3ss And T2dm · 4.2. T3ss And Gout
  78. [78]
    Type III secretion systems: the bacterial flagellum and the injectisome
    The flagellum and the injectisome are two of the most complex and fascinating bacterial nanomachines. At their core, they share a type III secretion system ...
  79. [79]
    Evolution of the type III secretion system and its effectors in plant ...
    Dec 6, 2007 · We explore the evolutionary origins of the T3SS relative to the closely related flagellar system, and investigate the evolutionary pressures on ...
  80. [80]
  81. [81]
    Bacterial type III secretion systems are ancient and evolved by ...
    Our analysis indicates that the TTSS and the flagellar export mechanism share a common ancestor, but have evolved independently from one another. The suggestion ...Missing: system orthologs
  82. [82]
    Molecular mechanisms of bacterial virulence: type III secretion ... - NIH
    The genes encoding several type III secretion systems reside on pathogenicity islands, which are inserted DNA segments within the chromosome that confer ...
  83. [83]
    Type III secretion systems in symbiotic adaptation of pathogenic and ...
    In pathogens, the type III secretion system is used to inject multiple effectors into host cells and helps Salmonella to colonize plant and animal hosts [17,24] ...
  84. [84]
    A global survey of bacterial type III secretion systems and their ...
    Apr 12, 2017 · Through a comprehensive survey of ∼20 000 bacterial genomes, we identified 174 non-redundant T3SSs from 109 genera and 5 phyla. Many of the ...Missing: prevalence | Show results with:prevalence
  85. [85]
    Computational prediction shines light on type III secretion origins
    Oct 7, 2016 · Type III secretion system is a key bacterial symbiosis and pathogenicity mechanism responsible for a variety of infectious diseases, ...
  86. [86]
    Prediction of Type III Secretion Signals in Genomes of Gram ...
    We present a signal prediction method together with comprehensive survey of potential type III secretion system effectors extracted from 918 published bacterial ...<|control11|><|separator|>
  87. [87]
    T3_MM: A Markov Model Effectively Classifies Bacterial Type III ...
    This research focused on comparing the amino acid composition within the N-terminal 100 amino acids from type III secretion (T3S) signal sequences or non-T3S ...
  88. [88]
    High-accuracy prediction of bacterial type III secreted effectors ...
    We demonstrated that the BPBAac model outperformed other implementations in classification of T3S and non-T3S proteins, giving an average sensitivity of ∼90.97% ...
  89. [89]
    The type III secretion system translocases IpaC, SipC, and BipC are ...
    Likewise, a template-based homology modeling showed that BipC folds into a tertiary structure with long alpha helical bundles [17]. Thus, prior to our ...
  90. [90]
    T3DB: an integrated database for bacterial type III secretion system
    Apr 30, 2012 · Type III Secretion System (T3SS), which plays important roles in pathogenesis or symbiosis, is widely expressed in a variety of gram negative ...
  91. [91]
    DeepT3 2.0: improving type III secreted effector predictions by an ...
    Oct 4, 2021 · Of all the methods tested, Effective T3 achieved the lowest accuracy, precision and MCC metrics. Collectively, the above results demonstrate ...
  92. [92]
    Targeting bacterial pathogenesis by inhibiting virulence-associated ...
    Here, we review previously and recently identified inhibitors of virulence-associated bacterial secretions systems and discuss their potential as therapeutics.
  93. [93]
    Research Progress on Small Molecular Inhibitors of the Type 3 ...
    Another derivative INP0400 also inhibited the T3SS of Chlamydia, but by a different mechanism. Muschiol et al. [88] found that intracellular duplication and ...
  94. [94]
    Emerging antimicrobial therapies for Gram-negative infections in ...
    Feb 27, 2025 · A further anti-virulence strategy that has progressed to clinical trials focuses on blocking the action of the P. aeruginosa Type 3 Secretion ...
  95. [95]
    Small molecule inhibitors of type III secretion in Yersinia block the ...
    Feb 20, 2007 · Further screening resulted in INP0400 that blocked T3SS mediated virulence in Y. pseudotuberculosis as well as formation of inclusions both for ...
  96. [96]
    Antibodies Inhibiting the Type III Secretion System of Gram-Negative ...
    The T3SS is becoming an important anti-virulence target for many reasons. The T3SS is specific to Gram-negative pathogens, meaning any interventions targeting ...
  97. [97]
    Direct Neutralization of Type III Effector Translocation by the ...
    Monoclonal antibodies. Anti-LcrV MAbs 7.3 and 29.3 (both IgG1) were produced and provided by Jim Hill (Defence Science and Technology Laboratory [DSTL]).