Helminth parasites have evolved sophisticated methods for manipulating the host immune response to ensure long-term survival in their chosen niche, for example, by secreting products that interfere with the host cytokine network. Studies on the secretions of Heligmosomoides polygyrus have identified a family of transforming growth factor-β (TGF-β) mimics (TGMs), which bear no primary amino acid sequence similarity to mammalian TGF-β, but functionally replicate or antagonise TGF-β effects in restricted cell types. The prototypic member, TGM1, induces in vitro differentiation of Foxp3+ T regulatory cells and attenuates airway allergic and intestinal inflammation in animal models. TGM1 is one of a family of ten TGM proteins expressed by H. polygyrus. It is a five-domain modular protein in which domains 1–2 bind TGFBR1, and domain 3 binds TGFBR2; domains 4–5 increase its potency by binding a co-receptor, CD44, highly expressed on immune cells. Domains 4–5 are more diverse in other TGMs, which bind co-receptors on cells such as fibroblasts. One variant, TGM6, lacks domains 1–2 and hence cannot transduce a signal but binds TGFBR2 through domain 3 and a co-receptor expressed on fibroblasts through domains 4–5 and blocks TGF-β signalling in fibroblasts and epithelial cells; T cells do not express the co-receptor and are not inhibited by TGM6. Hence, different family members have evolved to act as agonists or antagonists on various cell types. TGMs, which function by molecularly mimicking binding of the host cytokine to the host TGF-β receptors, are examples of highly evolved immunomodulators from parasites, including those that block interleukin (IL)-13 and IL-33 signalling, modulate macrophage and dendritic cell responses and modify host cell metabolism. The emerging panoply and potency of helminth evasion molecules illustrates the range of strategies in play to maintain long-term infections in the mammalian host.

Pathogens that establish chronic infections in their host must first evade the immune responses designed to lyse, kill, incapacitate, or eject them. Micro-organisms can take the approach of outpacing immunity and undergo rapid selection of escape mutants, while protozoal parasites can vary their surface-exposed antigens, such as with trypanosomes [1,2], rendering the antibody response raised against them to be non-functional. This strategy is not available to slower growing helminth macro-parasites, which do not proliferate in their hosts and, therefore, cannot rapidly select for advantageous mutations. Hence, many parasites take alternative routes through transition into different life stages and/or by releasing immunomodulatory factors, which tailor the immune response to their benefit [3]. Such sophisticated immunomodulation can result in a tolerance to the parasite, which at best can benefit both the parasite and host by minimising collateral damage, immune hyper-responsiveness and enabling asymptomatic chronic infection. More detrimentally, helminth infection can result in immune suppression, co- or hyper-infection, cancer progression/metastasis and reduced vaccination protection against other pathogens [4-6].

In viral infections, immune evasion factors are well understood and shown to suppress critical pathways, such as major histocompatibility complex class I antigen presentation at virtually every step in the process. In contrast, helminth immunomodulation has only begun to be understood with the recent expansion of molecular studies on these parasites. A broad range of mediators have now been discovered [7-11], which either block anti-helminth immune responses or induce immune tolerance depending on their life stage or tissue residence.

Cytokines are soluble messenger proteins for intercellular communication that activate and regulate various aspects of immunity, inflammation and tissue homeostasis [12-15]. The complexity of the cytokine network reflects the diverse nature of pathogens, each requiring a tailored immune response. Interestingly, redundancy within this network may have arisen from pathogens attempting to disrupt cytokine functions. This review focuses not on the role of cytokines during an infection but rather on how parasites alter the host cytokine network to their benefit.

A particularly intriguing study area is the hijacking of transforming growth factor-β (TGF-β) signalling by secreted parasite cytokines, exploiting host cell surface TGF-β serine/threonine kinase receptors (TGFBR1 and TGFBR2). TGF-β is an exceptional cytokine with functions far beyond the immune system, playing crucial roles in development, cellular metabolism, tissue homeostasis and repair [16,17]. Within the immune system, TGF-β is largely viewed as an anti-inflammatory cytokine through its induction of regulatory T cells, but in parallel, it acts to drive class switching and differentiation of IgA+ B cells and modifies dendritic cell (DC) and macrophage phenotypes [18]. The critical nature of TGF-β-mediated immune regulation is highlighted by the early mortality of mice lacking TGF-β1, through the development of multi-organ inflammatory disease, particularly in the intestinal tract [19]. Due to the critical role of TGF-β signalling in tissue differentiation and formation, mice lacking the other isoforms (TGF-β2 and TGF-β3) show congenital developmental defects [20,21], and animals deficient in TGF-β receptor II (TGFBR2) suffer embryonic or perinatal mortality [19].

While TGF-β is critical for survival in mammals, it poses threats — especially in cancer [22]. The tumour microenvironment often contains elevated levels of TGF-β, which has been found to block T cell activation during checkpoint immunotherapy [23,24]. As a potent immune suppressor, TGF-β is also detrimental in various infectious settings [25], particularly in parasitic infections. This was first reported in the moderation of T cell responses to the river blindness nematode Onchocerca volvulus [26], and in the high levels of serum TGF-β in patients with Chagas’ disease, caused by the protozoan Trypanosoma cruzi, linked to the development of myocardiopathy [27].

In experimental animal models, an evident role for TGF-β was revealed upon infection by the murine intestinal helminth Heligmosomoides polygyrus, in which administration of the small molecule inhibitor SB431542 (that blocks the kinase activity of TGFBR1 and closely related receptors) accelerated worm expulsion [28]. Parallel results were obtained using the neutralising anti-TGF-β monoclonal antibody 1D11 in mice, significantly reducing infection with another intestinal parasite Trichuris muris [29]. H. polygyrus infection also elicits in vivo expansion of regulatory T cells that are required for parasite persistence [30]. The ability of H. polygyrus to stimulate Tregs suggested that it may activate host TGF-β signalling, a proposition that was confirmed when secretions collected from the parasite in vitro (H. polygyrus excretory/secretory products, HES) were added to T cells and induced expression of transcription factor Foxp3. Induction was found to be inhibited by SB431542 [28], confirming that the parasite could directly address the TGF-β pathway. Similar TGF-β-like activity has been reported in excretory/secretory (ES) products of a related Trichοstrongylid nematode, Teladorsagia circumcincta [28] and in extracts of a rodent filarial nematode Litomosoides sigmodontis [31], but only in the case of H. polygyrus has the active principle been identified as described below.

Since HES is a complex mixture of many hundred soluble proteins released by H. polygyrus, this material was fractionated by ion exchange and size exclusion chromatography, and fractions were screened for TGF-β-like activity. Proteins in active fractions were identified by mass spectrometry reconciled to a transcriptomic database from different stages of the parasite [32,33]. Selected candidates were expressed in a mammalian cell system as recombinant proteins, one of which proved to be a highly potent activator of TGF-β-like signalling, both in triggering an intracellular effector SMAD3-dependent reporter in fibroblasts, and by the induction of Foxp3 in murine T cells [34]. Remarkably, this active protein does not share primary amino acid sequence similarity with any TGF-β family member, which includes three TGF-β isoforms, activins and bone morphogenetic proteins [35]. It is, however, distantly related to the Complement Control Protein (CCP) family, which, although barely apparent at the amino acid sequence level (Figure 1a), is confirmed by conservation of three-dimensional structure (Figure 1b) [36]. Interestingly, the parasite product has acquired several insertions of amino acid tracts that enlarge external loops compared with canonical domains, such as human complement receptor CD46 (Figure 1b).

Sequence and structural characteristics of TGM1 and CCP proteins.

Figure 1:
Sequence and structural characteristics of TGM1 and CCP proteins.

(a) Alignment of TGM1 D3 with domain 2 of human CD46 [36]. Identical residues are shown in inverse shading, similar residues in shaded boxes. Amino acid positions in the respective proteins are indicated above and below the alignment. (b) Structural comparison of TGM1 D3 [36], left, with domain 2 of human CD46 (PDB 1CKL), centre and overlay of both structures, right. N- and C-termini of the domains are indicated, and positions of key amino acids. The four β sheets are indicated, as is the α1 helis. HVL, Hyper-Variable Loop. This part of the figure is reproduced from Ref. 36 in the Journal of Biological Chemistry, in accordance with the republishing policy of the American Society for Biochemistry and Molecular Biology. (c) Alpha fold model of TGM1, indicating receptor specificities of the different domains, and the experimentally determined dissociation constants (Kd) for TGFBR1 and TGFBR2. Affinities of TGM4 and TGM6 for these receptors are given in Table 1.

Figure 1:
Sequence and structural characteristics of TGM1 and CCP proteins.

(a) Alignment of TGM1 D3 with domain 2 of human CD46 [36]. Identical residues are shown in inverse shading, similar residues in shaded boxes. Amino acid positions in the respective proteins are indicated above and below the alignment. (b) Structural comparison of TGM1 D3 [36], left, with domain 2 of human CD46 (PDB 1CKL), centre and overlay of both structures, right. N- and C-termini of the domains are indicated, and positions of key amino acids. The four β sheets are indicated, as is the α1 helis. HVL, Hyper-Variable Loop. This part of the figure is reproduced from Ref. 36 in the Journal of Biological Chemistry, in accordance with the republishing policy of the American Society for Biochemistry and Molecular Biology. (c) Alpha fold model of TGM1, indicating receptor specificities of the different domains, and the experimentally determined dissociation constants (Kd) for TGFBR1 and TGFBR2. Affinities of TGM4 and TGM6 for these receptors are given in Table 1.

Close modal

Importantly, unlike the mature bioactive TGF-β protein – which consists only of the ~110 aa C-terminal domain of a longer precursor – the H. polygyrus protein contains five homologous but nonidentical domains each forming a self-contained module, connected by short connecting segments (Figure 1c). A further striking distinction is that TGF-β is synthesised as an inactive pre-protein and stored until activated by integrins or a proteolytic event and released from latency-associated partners [37,38]. In contrast, the helminth product is immediately competent as a full-length native protein, and it could be argued that the parasite needs no such restraint on the activity of its immune modulator. Thus, it was named TGM1 indicating that it mimics TGF-β without being an evolutionarily related homologue. As detailed below, the domains act as modular units, each conferring separate functional properties to the full-length protein.

TGM1 replicates the functional effects of TGF-β across a range of immune settings (Figure 2). For example, both murine [39] and human [40] T cells respond to TGM1 by inducing Foxp3 expression specifying Treg function. A closer comparison of the dose responses to the two ligands shows that while TGF-β activates fibroblasts at lower concentrations than TGM1, the latter is more potent than TGF-β at higher levels, displaying a steeper slope indicative of co-operativity in binding [39]. Similarly, SMAD2/3 phosphorylation in T cells is more rapid in response to TGF-β, but the slower response to TGM1 is sustained for longer [39]. As will be discussed below, the discovery of diverse co-receptor interactions with TGM1, but not TGF-β, may provide a mechanistic explanation for these observations.

Immunological functions of TGM1 in vitro and in vivo.

Figure 2:
Immunological functions of TGM1 in vitro and in vivo.

Schematic representing the various immune functions of TGM1. Clockwise from top left. (a) Induction of Foxp3 expression in murine [34,39 ] and human [40] T cells, representing the regulatory T cell (Treg) phenotype. (b) In vivo suppression of experimental autoimmune encephalomyelitis by adoptively transferred Tregs generated by in vitro treatment with TGM1, compared with TGF-β-induced Tregs [39]. (c) Reduction in cardiac injury during myocardial infarction by intravenous (i.v.) administration of TGM1 [39]. (d) Acceleration of skin wound healing in mice by topical application of TGM1 [41]. (e) Amelioration of colitis in RAG-deficient mice receiving Foxp3-negative T cells, in animals given subcutaneous osmotic minipumps releasing TGM1 [42]. (f) Suppression of allergic airway eosinophilic inflammation in bronchoalveolar lavage fluid (BALF) in mice receiving intranasal Alternaria alternata fungal allergen, and treated with intraperitoneal (i.p.) TGM1 [43]. (g) Inhibition of LPS-driven IL-6 release from murine bone marrow-derived macrophages in the presence of TGM1 [44]. TGF, transforming growth factor; TGM, transforming growth factor-β mimics.

Figure 2:
Immunological functions of TGM1 in vitro and in vivo.

Schematic representing the various immune functions of TGM1. Clockwise from top left. (a) Induction of Foxp3 expression in murine [34,39 ] and human [40] T cells, representing the regulatory T cell (Treg) phenotype. (b) In vivo suppression of experimental autoimmune encephalomyelitis by adoptively transferred Tregs generated by in vitro treatment with TGM1, compared with TGF-β-induced Tregs [39]. (c) Reduction in cardiac injury during myocardial infarction by intravenous (i.v.) administration of TGM1 [39]. (d) Acceleration of skin wound healing in mice by topical application of TGM1 [41]. (e) Amelioration of colitis in RAG-deficient mice receiving Foxp3-negative T cells, in animals given subcutaneous osmotic minipumps releasing TGM1 [42]. (f) Suppression of allergic airway eosinophilic inflammation in bronchoalveolar lavage fluid (BALF) in mice receiving intranasal Alternaria alternata fungal allergen, and treated with intraperitoneal (i.p.) TGM1 [43]. (g) Inhibition of LPS-driven IL-6 release from murine bone marrow-derived macrophages in the presence of TGM1 [44]. TGF, transforming growth factor; TGM, transforming growth factor-β mimics.

Close modal

Although Foxp3+ Tregs induced by TGF-β signalling are functionally immunosuppressive, they may not maintain this phenotype and can revert to a Foxp3- effector phenotype, especially in an inflammatory environment [45]. However, a greater proportion of cells activated by TGM1 retain Foxp3 expression after transfer into mice with intestinal inflammation induced by dextran sodium sulphate, than is the case for TGF-β-induced Tregs [39]. TGM1 has a greater capacity to convert human Th17 effectors into Foxp3+ Tregs, arguing that memory as well as naïve T cells are converted into the regulatory compartment [40]. TGM1-induced Tregs are also fully functional with equivalent potency to TGF-β-induced Tregs when transferred to mice in an experimental autoimmune encephalomyelitis model [39]. Additionally, soluble TGM1 protein administered intraperitoneally protects mice against both airway allergy [43] and inflammatory colitis [42], and in the latter model, oral administration of TGM1 also attenuated disease [46], although in the in vivo setting, TGF-β signalling is likely to activate multiple cell types in addition to Tregs. In other analyses, TGM1 given intravenously reduced heart damage and scarring following myocardial infarction in mice [47], while when applied to skin wounds, it accelerated wound repair with a qualitative shift to scar-free healing that even renewed hair follicle formation [41].

Further investigation of the H. polygyrus secretome characterised by proteomic analysis of HES products [32,33] revealed a larger family of ten TGF-β mimic (TGM) proteins [48], (Table 1) comprising a variable number of homologous but non-identical domains (Figure 3a). Like other proteins, each domain contains at least two disulphide bonds that are fully conserved across all domains of each TGM family member; within the H. polygyrus genome, domains are each encoded by two exons with boundaries coinciding with domain junctions, indicative of an evolutionary module that can be duplicated and reduplicated over time. Indeed, a phylogenetic tree (Figure 3b) suggests that the three-domain proteins, TGM6, 9 and 10, are ancestral to the longer homologues, in line with earlier work indicating that domain (D)5 of TGM-10 may be the most primordial domain in the family [51]. Of note, TGM1–6 are predominantly produced by adult worms, and TGM7–10 by the tissue-dwelling larvae [33,52].

Table 1:
Features of TGM family members.
 NCBIMFB-F11 SMADT cell Foxp3 TGFBR1TGFBR2Co-receptor(s)References
TGM1 422 aa SP + 5 domains MG099712 AT059092 +++ +++ 90 ± 1 nM 610 ± 10 nM CD44 30 nM [34,48
TGM2 430 aa SP + 5 domains MG429737 AVN88293 +++ +++ (100% identical to TGM-1)   [48] Ntang, E.Y.1 
TGM3 429 aa SP + 5 domains MG429738 AVN88294 +++ +++ (100% identical to TGM-1) (100% identical to TGM-1)  [48
TGM4 422 aa SP + 5 domains MG429739 AVN88295 – ++ 5.0 ± 0.1 nM 116 ± 2 µM CD44 20 nM, CD49d CD206 [48,49
TGM6 254 aa SP + 3 domains MG429741 AVN88297 – – N/A (D1-2 are absent) 220 ± 100 nM TBD : Not CD44 [48,50
TGM7 599 aa SP + 7 domains MG429742 AVN88298 – – Not tested Not tested TBD : Not CD44 [48
TGM9 251 aa SP + 3 domains MG429744 AVN88300 – Not tested N/A (D1-2 are absent) Not tested TBD : Not CD44 Mullin, T. and Singh, S.P.1 
TGM10 251 aa SP + 3 domains MG429745 AVN88301 – Not tested N/A (D1-2 are absent) Not tested  Ciancia, C.1 
 NCBIMFB-F11 SMADT cell Foxp3 TGFBR1TGFBR2Co-receptor(s)References
TGM1 422 aa SP + 5 domains MG099712 AT059092 +++ +++ 90 ± 1 nM 610 ± 10 nM CD44 30 nM [34,48
TGM2 430 aa SP + 5 domains MG429737 AVN88293 +++ +++ (100% identical to TGM-1)   [48] Ntang, E.Y.1 
TGM3 429 aa SP + 5 domains MG429738 AVN88294 +++ +++ (100% identical to TGM-1) (100% identical to TGM-1)  [48
TGM4 422 aa SP + 5 domains MG429739 AVN88295 – ++ 5.0 ± 0.1 nM 116 ± 2 µM CD44 20 nM, CD49d CD206 [48,49
TGM6 254 aa SP + 3 domains MG429741 AVN88297 – – N/A (D1-2 are absent) 220 ± 100 nM TBD : Not CD44 [48,50
TGM7 599 aa SP + 7 domains MG429742 AVN88298 – – Not tested Not tested TBD : Not CD44 [48
TGM9 251 aa SP + 3 domains MG429744 AVN88300 – Not tested N/A (D1-2 are absent) Not tested TBD : Not CD44 Mullin, T. and Singh, S.P.1 
TGM10 251 aa SP + 3 domains MG429745 AVN88301 – Not tested N/A (D1-2 are absent) Not tested  Ciancia, C.1 

TGM5 and TGM8 are not included as they have not been expressed or tested in experimental settings so far.

1

Unpublished studies.

SP+, predicted signal peptide (aa 1-18). TBD, to be determined.

TGM family member domain organisation and phylogenetic relationship.

Figure 3:
TGM family member domain organisation and phylogenetic relationship.

(a) (Top section) Domain organisation of TGM1 and corresponding 11 exons in the H. polygyrus genome (Roman numerals); yellow C letters in black circles denote Cys residues. All disulphide bonds are intra-domain. (Main section) Organisation of domains in 10 family members, with percent amino acid identity of each domain to the corresponding domain of TGM1. Δ denotes absence of the domain(s). Note TGM7 and TGM8 have two additional domains, percentage identities shown are as indicated by colouring, with those with D4 of TGM1. Schematic is updated from Smyth et al [48] with corrected percent identities for D4-5 of TGM2 and the domains of TGM10 realigned. (b) Phylogenetic tree of the 10 full-length sequences of TGM family members. Numerals represent P values for branching topography. NB: For clarity, the TGM proteins are shown hyphenated (TGM-1), although normally the hyphen is omitted. TGF, transforming growth factor; TGM, transforming growth factor-β mimics.

Figure 3:
TGM family member domain organisation and phylogenetic relationship.

(a) (Top section) Domain organisation of TGM1 and corresponding 11 exons in the H. polygyrus genome (Roman numerals); yellow C letters in black circles denote Cys residues. All disulphide bonds are intra-domain. (Main section) Organisation of domains in 10 family members, with percent amino acid identity of each domain to the corresponding domain of TGM1. Δ denotes absence of the domain(s). Note TGM7 and TGM8 have two additional domains, percentage identities shown are as indicated by colouring, with those with D4 of TGM1. Schematic is updated from Smyth et al [48] with corrected percent identities for D4-5 of TGM2 and the domains of TGM10 realigned. (b) Phylogenetic tree of the 10 full-length sequences of TGM family members. Numerals represent P values for branching topography. NB: For clarity, the TGM proteins are shown hyphenated (TGM-1), although normally the hyphen is omitted. TGF, transforming growth factor; TGM, transforming growth factor-β mimics.

Close modal

A functional analysis of TGM1 in which amino (N)- and carboxy (C)-terminal domains were deleted showed that the minimal structure required for signalling through the TGF-β pathway comprises D1-3 [48]. Biophysical studies by surface plasmon resonance (SPR) and isothermal titration calorimetry established that D1-2 bind TGF-β receptor subunit I (TGFBR1) and D3 binds TGFBR2 [36] (Figure 1c). Mapping of the binding sites on TGFBR1 and TGFBR2 using nuclear magnetic resonance [36] further showed that D1-2 and D3 engage similar structural features and residues on these receptors as does the native ligand, TGF-β. Recently, a high-resolution (1.4 Å) structure of the D3-TGFBR2 complex (Figure 4a) revealed the remarkable mimicry of the binding interactions as compared with mammalian TGF-β, in spite of an entirely different structural framework (Figure 4b).

Mimicry of TGFBR2 binding by TGM domain 3.

Figure 4:
Mimicry of TGFBR2 binding by TGM domain 3.

(a) Structure of TGM6-D3 (magenta) bound to the human TGFBR2 ectodomain (blue). Structure was determined using X-ray crystallography and was determined to a resolution of 1.4 Å [50]. Central hydrophobic interaction in which TGFBR2 Ile76 inserts into a pocket formed by TGM6-D3 Ile78, Tyr80 and Tyr93 is highlighted by yellow shading. Peripheral hydrogen-bonded ion pairs, TGFBR2 Asp55 and Glu78 with TGM6-D3 Arg95 and Arg38, respectively, and TGFBR2 Asp141 and Glu142 with TGM6-D3 Tyr80 and Arg82, respectively, that further stabilise the interaction and add specificity are highlighted by blue shading. (b) Structure of TGF-β3 (olive) bound to the human TGFBR2 ectodomain (blue). Structure was determined using X-ray crystallography and was determined to a resolution of 3.0 Å [53]. Central hydrophobic interaction in which TGFBR2 Ile76 inserts into a pocket formed by TGF-β3 Trp332, Tyr390 and Val392 is highlighted in yellow. Peripheral hydrogen-bonded ion pairs, TGFBR2 Asp55 with TGF-β3 Arg394 and TGFBR2 Glu142 with TGF-β3 Arg325, that further stabilise the interaction and add specificity are highlighted in yellow. TGF, transforming growth factor; TGM, transforming growth factor-β mimics.

Figure 4:
Mimicry of TGFBR2 binding by TGM domain 3.

(a) Structure of TGM6-D3 (magenta) bound to the human TGFBR2 ectodomain (blue). Structure was determined using X-ray crystallography and was determined to a resolution of 1.4 Å [50]. Central hydrophobic interaction in which TGFBR2 Ile76 inserts into a pocket formed by TGM6-D3 Ile78, Tyr80 and Tyr93 is highlighted by yellow shading. Peripheral hydrogen-bonded ion pairs, TGFBR2 Asp55 and Glu78 with TGM6-D3 Arg95 and Arg38, respectively, and TGFBR2 Asp141 and Glu142 with TGM6-D3 Tyr80 and Arg82, respectively, that further stabilise the interaction and add specificity are highlighted by blue shading. (b) Structure of TGF-β3 (olive) bound to the human TGFBR2 ectodomain (blue). Structure was determined using X-ray crystallography and was determined to a resolution of 3.0 Å [53]. Central hydrophobic interaction in which TGFBR2 Ile76 inserts into a pocket formed by TGF-β3 Trp332, Tyr390 and Val392 is highlighted in yellow. Peripheral hydrogen-bonded ion pairs, TGFBR2 Asp55 with TGF-β3 Arg394 and TGFBR2 Glu142 with TGF-β3 Arg325, that further stabilise the interaction and add specificity are highlighted in yellow. TGF, transforming growth factor; TGM, transforming growth factor-β mimics.

Close modal

While domains D1-3 bind to the canonical receptors that are required for signalling, the function of D4 and D5 has only recently been clarified. Our findings indicated that TGM1 lacking D4 and D5 is considerably less potent than the full-length TGM1, suggesting that D4-5 are necessary for achieving the full potency of TGM1. This led us to speculate that D4 and D5 mediate cell specificity and potency by interacting with co-receptors. Indeed, TGM1 D4 and 5 have been found to interact with cell adhesion receptor CD44, which has a cell-type-restricted expression pattern and is essential for the potency of TGM1 [54] (Figure 1c). CD44 is an adhesion and migration molecule [55,56] widely expressed on immune cells and up-regulated on key subsets of memory/effector T cells [57], but is also found on endothelial and epithelial cells. In the context of immune cells, CD44 is required for efficient TGM1 activation, as shown both by reduced responses of CD44-deficient cells, and the loss of potency when D4-5 are removed from TGM1 and the remaining D1-3 is tested on fibroblasts or T cells [54]. Hence, it is suggested that TGM1 may have evolved to target CD44-expressing immune cells to dampen the host immune system during H. polygyrus infection; whether it also modulates epithelial cells such as CD44+ intestinal stem cells [58] remains to be determined.

Mammalian TGF-β homodimers have a compact structure that interacts with ubiquitously expressed TGFBR1 and TGFBR2 to assemble TGFBR12:TGFBR22 heterotetramers [59]. In contrast, TGM1 engages with TGFBR1, TGFBR2, through two independent modules (D1-2, D3) to form a TGFBR1:TGFBR2 heterodimer, and in association with a co-receptor through a third module (D4-5). For the biological activity induced by TGF-β1 and TGF-β3, the presence of TGFBR1 and TGFBR2 alone is sufficient. However, for TGF-β2, both TGFBR1 and TGFBR2 and the TGFBR3/betaglycan co-receptor are required [60,61]. TGFBR3 primes cells to respond to TGF-β2 by potentiating receptor complex assembly and signalling and is a key determinant for TGF-β2 cell specificity [44,62]. The way TGM1 binds to cell surfaces is, therefore somewhat similar to TGF-β2, as signalling requires the cooperation of three receptors. However, unlike TGF-β2 where the co-receptor TGFBR3 is displaced by the signalling receptors [44,63], TGM1 independently binds TGFBR1 through D1-2, TGFBR2 through D3 and remains bound to the co-receptor through D4-5 as it signals through TGFBR1 and TGFBR2.

Of the family of ten TGMs, TGM1–4 share a similar modular structure with five discrete domains, while TGM5, TGM6, TGM9 and TGM10 lack one or two domains, and TGM7 and TGM8 have acquired two additional domains (Figure 3a). The D1-3 of TGM1 that bind to TGFBR1 and TGFBR2 share high amino acid sequence identity (90–100%) with the corresponding domains of TGM2, 3, 4 and 5 but have lower sequence identity (50–66%) with those of TGM6–10. At the biophysical level, D3 of TGM1, 4 and 6 has each been shown to interact with TGFBR2 [36,49,50], and this suggests that all TGMs that include D3 may interact with TGFBR2, and those with D12 with TGFBR1.

The D4-5 of TGM1 are similar (87–99%) to D4-5 of TGM2, 3, 4 and 5 and are likely to share the same co-receptors. Indeed, we confirmed experimentally that TGM1 and TGM4 share CD44 as a co-receptor partner [49,54], but distinct from TGM1, TGM4 also interacts with CD49d (integrin α4) and CD206. Co-receptors confer cell specificity for TGF-β signalling as the individual affinity of TGMs for the TGFBRs can be moderate to low, such that the D3 of TGM1 and TGM4 binds TGFBR2 with affinities in the range of 1–2 and 50–100 μM, respectively [36,49]. The broader range of cell surface co-receptors for TGM4 compared with TGM1 results in increased cell specificity of TGM4 for monocytes rather than T lymphocytes or fibroblasts [49], prescribing a myeloid cell preference for TGM4 that may allow it to dampen immune activation without incurring the risk of fibrosis (Figure 5).

Model of cell specific actions of TGM proteins.

Figure 5:
Model of cell specific actions of TGM proteins.

Conceptual model comparing receptor interactions and cell specificity of TGF-β, TGM4 and TGM6. Created in BioRender. McSorley, HJ. (2025) https://BioRender.com/a55n320 (a) TGF-β acts as a homodimer to assemble a heterotetrameric complex of TGFBR12:TGFBR22 that signal to all cells expressing these receptors. (b) TGM4 ligates co-receptors including CD44, CD49d and CD206, while directly binding TGFBR1 and TGFBR2. D4-5 of TGM4 bind to CD44 and CD206; the domain binding to CD49d has not been conclusively identified. As a result of co-receptor interactions, TGM4 is selectively active on immune cells, particularly macrophages and does not activate fibroblasts with low or no expression of these co-receptors. (c) TGM6 lacks D1-2 and so cannot bind TGFBR1; it does however bind TGFBR2 together with a co-receptor (Co-R) that is different from those identified binding to TGM1 or TGM4. When added to cells expressing the appropriate co-receptor, such as fibroblasts, TGM6 acts as an antagonist of TGF-β signalling. TGF, transforming growth factor; TGM, transforming growth factor-β mimics.

Figure 5:
Model of cell specific actions of TGM proteins.

Conceptual model comparing receptor interactions and cell specificity of TGF-β, TGM4 and TGM6. Created in BioRender. McSorley, HJ. (2025) https://BioRender.com/a55n320 (a) TGF-β acts as a homodimer to assemble a heterotetrameric complex of TGFBR12:TGFBR22 that signal to all cells expressing these receptors. (b) TGM4 ligates co-receptors including CD44, CD49d and CD206, while directly binding TGFBR1 and TGFBR2. D4-5 of TGM4 bind to CD44 and CD206; the domain binding to CD49d has not been conclusively identified. As a result of co-receptor interactions, TGM4 is selectively active on immune cells, particularly macrophages and does not activate fibroblasts with low or no expression of these co-receptors. (c) TGM6 lacks D1-2 and so cannot bind TGFBR1; it does however bind TGFBR2 together with a co-receptor (Co-R) that is different from those identified binding to TGM1 or TGM4. When added to cells expressing the appropriate co-receptor, such as fibroblasts, TGM6 acts as an antagonist of TGF-β signalling. TGF, transforming growth factor; TGM, transforming growth factor-β mimics.

Close modal

TGM6, 9 and 10 stand out for their absence of D1-2 which, in other family members, bind TGFBR1. We hypothesised that these proteins could act as antagonists if, for example, they sequestered TGFBR2 without recruiting TGFBR1to permit its phosphorylation. Indeed, TGM6 was recently found to retain and even have enhanced binding to TGFBR2 compared with TGM1 (Kd 1-2 μM and ~0.3 μM for TGM1 and TGM6, respectively) [36,50]. TGM6 also competed with TGF-β for binding TGFBR2, but it did not bind TGFBR1 or any other type I receptors of the TGF-β family. In reporter assays in mouse cells, such as MFB-F11 and NIH-3T3 fibroblasts, TGM6 was found to potently inhibit signalling by either TGM1 or TGF-β with sub-nanomolar potency [50]. However, TGM6 was unable to inhibit TGF-β signalling in some other cell types, such as splenic T cells. The latter, together with the inability of TGM6 to bind CD44, suggests that TGM6 may require a co-receptor, presumably engaged by D4-5, that is specific to fibroblasts and other populations such as epithelial cells that can also be inhibited by TGM6, in contrast with TGM4 which is specific for immune cells through a different co-receptor profile (Figure 5).

TGM6, 7, 8, 9 and 10 share lower sequence similarity (28–32%) with D4-5 of TGM1 and are likely to interact with different co-receptors than TGM1. Consistent with this notion, we found that TGM6 did not bind CD44 and antagonism by TGM6 depended on the physical attachment of D3 with D4-5 [50]. Our TGM (co)receptor identification studies, with both TGM agonists and antagonists, suggest that TGMs deliver TGF-β agonistic or antagonistic signals via in-cis interaction with ubiquitously expressed host TGF-β receptors and cell-type selective co-receptors. Therefore, identification of co-receptors for TGM6, 7, 8, 9 and 10 and the cell populations that express these receptors is eagerly awaited.

TGM1–10 are all produced by the same species, H. polygyrus. One possible reason for the secretion of multiple TGMs is that TGF-β is a highly contextual and multifunctional protein. Activating a TGM with similar binding and signalling characteristics to mammalian TGF-β might confer benefits, but it would also likely have adverse effects and thus offer no net benefit to H. polygyrus. To maximise their fecundity and longevity, the modular domain structure of the TGMs has enabled H. polygyrus to evolve the secretion of TGMs that specifically activate or inactivate TGF-β signalling in specific cell types (Figure 5). Our recent TGM binding studies have provided insights into how the cell-specific mechanism is achieved. The key point is that the affinities of TGMs D1-3 for signalling TGF-β receptors, particularly TGFBR2, are too weak to trigger TGF-β (agonistic) responses. For instance, TGM4 has more than a 100-fold lower affinity for TGFBR2 than TGF-β. Moreover, TGMs D1-3 must be physically connected to D4-5 (as they are in wildtype TGMs) to elicit TGF-β modulatory responses [36,49,50]. If cells are treated with a combination of TGM4–5 and truncated versions of TGM D1-3, TGM activity cannot be rescued. These findings suggest that binding TGM D4-5 to co-receptors is essential to increase cell surface avidity, above that accomplished solely by TGM D1-3 alone. The simultaneous combinatorial TGM interactions with (co-)receptors in-cis allow for the delivery of cell-specific specific responses.

H. polygyrus enhances its survival in mouse hosts by exploiting the immune suppressive pathway mediated by TGF-β. It achieves this by having convergently evolved the functionally similar TGMs, which, despite primary amino acid sequences and structures being completely unrelated to that of mammalian TGF-β, are nonetheless able to interact with host TGFBRs. Phylogenetic analysis (Figure 3b) suggests that the three domain TGMs, TGM6, 9 and 10, are the ancestors of the family [51], indicating that D3 and its ligation to TGFBR2 was an ancestral characteristic. If correct, this suggests that TGMs likely evolved to antagonise the TGF-β pathway, perhaps as a mechanism to minimise fibrotic damage to the host as the parasite progresses through its life cycle in which the larvae bore though the intestinal epithelium to encyst in the muscle, and after maturation into adults, bore once again through the epithelium to the lumen to reproduce and for eggs to be excreted in the faeces.

Helminths are common parasites in humans and other animals, creating a notable interest in the presence of TGF-β mimetic proteins beyond the Heligmosomidae family, which includes H. polygyrus and species of the Heligmosomum genus. Some evidence suggests an archaic precursor in the ruminant nematode Haemonchus contortus, which belongs to the sister family Trichostrongylidae, both within the Strongyloidea superfamily [51]. The lack of TGMs beyond H. polygyrus is caused by a paucity of full-length cDNA sequences and incomplete whole genome sequences for species within and beyond its own taxonomic family. It would be valuable to address this gap and investigate the presence of TGMs in species other than H. polygyrus.

The TGF-β family is well conserved: TGF-β encoding genes are identifiable across all metazoan organisms [64,65]. The model nematode Caenorhabditis elegans expresses five TGF-β family encoded genes, DAF-7, TIG-3, DBL-1, TIG-2 and UNC-129, the first two being most closely related to TGF-β [66,67]. C. elegans also possesses both type I (DAF-1, SMA-6) and type II (DAF-4) receptors, the latter even able to bind human BMP-2 and BMP-4 proteins [68]. In the best characterised system, the DAF4 receptor controls entry of C. elegans into arrested development, which is blocked in the presence of the TGF-β family ligand DAF-7 [69], while more recent studies have reported a remarkable involvement of DBL-1 and TIG-2 in the innate immune response of C. elegans to bacterial infection [67].

Subsequently, multiple TGF-β family members and their receptors have been identified across a wide range of parasitic nematodes [70-75], trematodes [76,77] and cestodes [78]. The presence of TGF-β family genes across parasitic and non-parasitic helminth species implies that these genes have a developmental role in all helminths. Interestingly, while DAF-7 prevents arrested development in C. elegans, the role of TGF-β signalling appears to be reversed in parasitic species [79]. Arrest is obligatory for larval parasitic stages until they take the opportunity to infect a new host, and in species such as the mosquito-borne Brugia malayi, expression of the TGF-β homologue TGH-2 is maximal in blood-stage microfilariae that await uptake by the arthropod vector before recommencing development [71,79]. More broadly, genetic knockdown of TGF-β family members in parasitic helminths has confirmed developmental requirements for many of these genes, with roles in trematodes in embryogenesis [76] and in the moulting and development of parasitic nematodes [75]. For example, a recombinant activin-like product of the liver fluke Fasciola hepatica enhanced parasite egg embryonation and parasite viability [80], while RNAi knockdown of the corresponding gene compromised the development and movement of newly excysted juvenile parasites [81].

Since this well-conserved family of proteins retains similarity to their mammalian counterparts, it would be logical to propose that gene duplication of parasite TGF-β family members, and evolution to bind to host TGF-β receptors could be an efficient mechanism of parasite immunomodulation [82]. Indeed, it has been reported that TGF-β homologues from Brugia malayi (Bm-TGH-2) [71] and Fasciola hepatica (FhTLM) [77] may signal via host TGF-β family receptors. However, when tested in T cell culture assays, no helminth homologue has been found to replicate the Foxp3+ regulatory T cell expansion activity of host TGF-β or TGM1 [78,81]. A TGF-β homologue from Echinococcus multilocularis (EmACT) has been reported that is only distantly related to human TGF-β, with 24% amino acid identity in the C-terminal receptor-binding region, albeit slightly higher similarity to activin. EmACT cannot directly induce Foxp3+ regulatory T cells in culture, but can amplify Foxp3 induction by low concentrations of recombinant TGF-β [78]. This effect is similar to that seen for mammalian activin A, which requires TGF-β and TGF-β-like receptor signalling to mediate its regulatory T cell-promoting effects [83].

Some unanswered questions remain about these interactions. For example, do endogenous TGF-β receptors in helminths recognise and respond to host-derived ligands, as suggested by a report that exposure of adult S. mansoni worms to human TGF-β induces a gene response that is blocked by knockdown of the parasite type II receptor [84]. Furthermore, the possibility that worm receptors can respond to the H. polygyrus TGMs remains to be explored.

Therefore, parasitic nematodes produce multiple TGF-β family proteins, and it is possible that some may be involved in modulation of the host immune response. Further investigations are required to identify which of these TGF-β family proteins are developmental, immunomodulatory or both and to ascertain their complementary TGF-β family receptors. More broadly, the presence of bona fide helminth TGF-β homologues poses the question of why H. polygyrus evolved the TGM family from a completely unrelated ancestor to carry out this immunomodulatory function.

The TGF-β pathway is clearly a rich seam for parasite immunomodulation, but it is far from the only cytokine signalling pathway modulated by parasite secreted factors [10]. Parasite products target many cytokines that either induce or suppress type 2 immune responses, including the interleukin (IL)-13, IL-33 and macrophage migration inhibitory factor (MIF) pathways.

IL-13 is a critical effector cytokine in the type 2 immune response: although pleiotropic, it is particularly effective on stromal cells, inducing goblet and tuft cell differentiation in the intestinal tract, together with smooth muscle hyperplasia. These cellular effects increase mucus secretion and peristalsis, mediating the ‘weep and sweep’ response for helminth ejection. In T. muris infection, IL-13 production is particularly key to resistance versus susceptibility [85], with IL-13-deficient mice on a resistant genetic background becoming highly susceptible to the parasite. Therefore, IL-13 seems a prime target for modulation by T. muris. Secretions of T. muris are dominated by a single protein, p43 [86], the gene for which is the tenth most highly expressed transcript in the adult parasite [87]. p43 binds directly to IL-13 and blocks the activity of this cytokine, inhibiting IL-13 responses during infection. Importantly, although p43 is not immunogenic during natural infection, vaccination with p43 in an alum adjuvant resulted in parasite ejection, thought to be due to the blockade of p43-mediated immunomodulation of IL-13 [86].

Responses to the alarmin cytokine IL-33 are also strongly implicated in immunity to helminths, with increased susceptibility of mice deficient in either IL-33 or the IL-33 receptor (ST2) seen in a wide range of helminth infections [4]. Therefore, as is the case for IL-13, the IL-33 pathway constitutes a crucial focus of helminth immunomodulation. H. polygyrus HES suppresses IL-33 responses in models of asthma [88,89] and in helminth infection [90] through down-regulation of ST2 transcription [89] and through inducing IL-1β release, which in turn suppresses production of IL-33 and IL-25 [90]. Finally, HES contains two families of proteins that interact directly with IL-33 or ST2, respectively. The H. polygyrus Alarmin Release Inhibitor (HpARI) family binds directly to IL-33 [91,92]. The HpARI family contains three members (HpARI1-3), of which HpARI1 and HpARI2 suppress responses to IL-33, while HpARI3 stabilises IL-33 and amplifies responses to the cytokine [93]. It is unclear why the parasite encodes proteins that have directly opposing functions; however, an explanation may stem from the finding that while HpARI1 and HpARI2 also bind to the extracellular matrix, HpARI3 does not [94], which could result in spatial or temporal separation of their effects in vivo. Finally, HES also contains the Binds Alarmin Receptor and Inhibits (HpBARI) family, containing HpBARI and HpBARI_Hom2 [95]. Both HpBARIs act similarly, binding with high affinity to ST2 and blocking interaction between the cytokine and its receptor. Thus, considerable evidence indicates that H. polygyrus applies significant resources to suppress IL-33 pathway, indicating the importance of this cytokine in resistance to the parasite.

Remarkably, HpARI and HpBARI have also both descended from an ancestral domain and are thus distantly related to the TGM family; it appears that within the H. polygyrus lineage, there has been repeated duplication and expansion of -like genes so that they are highly represented in the parasite genome [10]. Thus, while no direct homologues of the HpARIs or HpBARIs have been identified in other parasites (as is also the case for TGMs), there are multiple domains encoded in all helminth genomes and it appears likely that other parasites will have adapted these for the purposes of immune evasion in one manner or another [96].

The MIF pathway, as its name suggests, is important in the activation of macrophages. Its effects are highly context-dependent, activating macrophages in inflammatory settings but also having a role in the induction of type 2 immune responses via activation of a type 2 innate lymphoid cell (ILC2)-tuft cell pathway [97]. As host MIF expression is required for optimal anti-parasite immunity [98], it is surprising that many parasitic helminths secrete MIF homologues. Indeed, the MIF family is well conserved across many organisms, whether free-living or parasitic [99]. Several parasitic helminth MIFs show immunosuppressive properties via induction of anti-inflammatory macrophage activation (Brugia malayi Bm-MIF1/2 [100], induction of immunosuppressive cytokines (Strongyloides ratti Sra-MIF [101]) or regulatory T cell recruitment (Anisakis simplex As-MIF [102]). With an eye to the host agonism/antagonism via the TGM and HpARI families, it would be timely to revisit this work to test whether closely related MIF homologues are agonists versus antagonists and whether like the TGMs they modulate MIF signalling in a context-specific manner that would be optimal for the parasite.

Helminths also secrete many other molecules that heavily affect host immune responses and target towards myeloid immune cells, such as DCs and macrophages. A well-studied example is the enzyme ribonuclease, omega-1, secreted by eggs of Schistosoma mansoni [103]. Omega-1 primed DCs for Th2 polarisation [104,105] through binding of the mannose receptor via Lewis X glycan structures. Upon internalisation in DCs, protein synthesis was inhibited [106], while omega-1 also affected inflammasome-dependent IL-1-β in macrophages [107]. Experimental models showed that the administration of recombinant omega-1 in vivo targeted type 2 conventional DC (cDC2) and prevented allergic asthma by hampering DC migration [108]. This effect was dominant over the type 2 polarisation. Likewise, the fatty acid binding protein (FABP)1 from F. hepatica influenced DC function and T cell polarisation through the induction of thrombospondin-1 in DCs [109]. The molecule ES-62 is produced by the filarial nematode Acanthocheilonema viteae and has anti-inflammatory activities demonstrated in numerous models of inflammatory diseases [110]. ES-62 is a tetrameric glycoprotein, comprising of identical monomers of ~62 kDa and exerts its anti-inflammatory effect through a phosphorylcholine (PC)-containing moiety. ES-62 targets toll-like receptor (TLR)4 and affects downstream signalling leading to enhanced extracellular signal-regulated kinase (ERK)1/2 activation and DC hyporesponsiveness [111]. Apart from DCs, B cells are also targeted by secreted schistosome enzymes and molecules. For example, both recombinant glycoprotein antigen IPSE/α1, Sm14 and Thioredoxin-1 (SmTrx-1), induced IL-10-producing regulatory B cells, instrumental in driving regulatory T cells and immune evasion [112,113].

Interestingly, an increasing number of studies suggest that helminth-derived molecules impact metabolic activity and in that manner modify the function of specific immune cells, like macrophages. For example, it was suggested that recombinant omega-1 improved whole-body metabolic homeostasis [114,115]. Another class of molecules in this context form the cystatins, cysteine protease inhibitors which are produced by numerous helminth species, including A. viteae, Ascaris lumbricoides, S. japonicum and S. mansoni. Cystatin administration shows inhibitory effects in disease models [116-118]. This may be partially regulated by enhanced mevalonate and cholesterol biosynthesis pathways and immunomodulatory gene activity in DCs and macrophages, as demonstrated for cystatins secreted by A. lumbricoides [119]. An immunomodulating peptide secreted by Fasciola hepatica, named helminth defence molecule (FhHDM)1 was able to switch macrophage metabolism to oxidative phosphorylation fuelled by fatty acids and the induction of glutaminolysis. This was accompanied by decreased tumour necrosis factor and IL-6 production pointing to a role of switched immunometabolism in immune suppression [120]. Lastly, a recent study demonstrated that helminthic glutamate dehydrogenase secreted by H. polygyrus actively suppresses macrophage-mediated host defence through induction of prostaglandin E2 and suppression of type 2-promoting leukotrienes via 2-hydroxyglutarate (2-HG) [121]. Increased 2-HG is associated with reprogrammed cellular metabolism and is usually linked to hypoxia and mitochondrial dysfunction. 2-HG is grouped with other tricarboxylic acid cycle metabolites, such as acetyl-CoA, itaconate, succinate, fumarate, which are known for their capacity to alter both innate and adaptive immune cell systems through their ability to modify chromatin remodelling and DNA methylation, rewiring the epigenetic landscape of immune cells [122]. We are only at the beginning of understanding how such metabolic cues can induce long-term functional consequences for immune cells and how helminths have mastered the art of manipulating these processes through the secretion of enzymes and other interference mechanisms.

The example of the helminth TGMs is instructive both in scope – that parasites can evolve unprecedented ligands that target pivotal pathways in host immunity – and in strategy, for combinatorial receptor binding is an especially effective means of selectively acting on specific host cell populations. As well as specificity, multivalency bestows avidity that will always exceed the affinity of any individual ligand-receptor pair. The multi-domain TGMs extend this further with trivalent interactions, an example with few parallels in extracellular receptor biology.

There are many fascinating nuances to be gleaned from this system: for example, the moderation of affinity for the canonical TGF-β receptors is key to the success of TGMs, as higher affinity would obviate the requirement for co-receptors and ablate cell specificity. Trivalency enables high avidity to be attained while maintaining individual receptor binding at low-to-medium affinity levels. Furthermore, maximal affinity does not always translate into optimal efficacy, as demonstrated in the setting of agonistic monoclonal antibodies for checkpoint immunotherapy, which act by driving a signal rather than blockade [123].

The interconversion of agonists and antagonists by gain or loss of binding modules represents a further quintessential feature of the TGM family. Mimicry to overturn function, as in the case of the antagonists, is not unique: the plant pathogen Phytophthora Suppressor of RNA silencing 2 protein PSR2 comprises divergent LWY modules, one of which mimics a host plant phosphatase subunit and combines with the remaining subunits to redirect its specificity to new protein targets [124].

In conclusion, TGM modularity offers exciting opportunities for exploitation in the setting of TGF-β signalling: if we can recombine, redesign and repurpose agonists and antagonists to different target cells and tissues, introducing novel modules with new specificities and fine-tuning affinities for maximum discriminatory properties, we may realise the therapeutic potential of this remarkable gene family across a wide range of disease settings.

The authors declare that there are no competing interests associated with the manuscript.

This work was funded by awards to HJM, RMM and HHS from LONGFONDS|Accelerate as part of the AWWA project; the UK Medical Research Council to HJM (MR/S000593/1); the NIH through an RO1 (GM58670) and RO3 (AI53915) awarded to APH; and the Wellcome Trust through Investigator Awards awarded to RMM (Ref 219530) and HJM (Ref 221914) and a Wellcome Trust Discovery Award jointly held by all five authors (Ref 306173).

For the purpose of open access, the authors have applied a Creative Commons Attribution (CC BY) licence to any Author Accepted Manuscript version arising from this submission.

R.M.M., H.J.M., H.H.S., P.D., and A.P.H.: Writing–original draft and writing–review and editing.

CCP

complement control protein

TGF

transforming growth factor

TGMs

transforming growth factor-ß mimics

1
Finlay
,
B.B.
and
McFadden
,
G
. (
2006
)
Anti-immunology: evasion of the host immune system by bacterial and viral pathogens
.
Cell
124
,
767
782
https://doi.org/10.1016/j.cell.2006.01.034
2
Schmid-Hempel
,
P
. (
2008
)
Parasite immune evasion: a momentous molecular war. trends ecol evol.
.
23
,
318
326
https://doi.org/10.1016/j.tree.2008.02.011
3
Maizels
,
R.M.
and
Yazdanbakhsh
,
M
. (
2003
)
Regulation of the immune response by helminth parasites: cellular and molecular mechanisms
.
Nat. Rev. Immunol.
3
,
733
744
https://doi.org/10.1038/nri1183
4
McSorley
,
H.J.
and
Smyth
,
D.J
. (
2021
)
IL-33 : A central cytokine in helminth infections
.
Semin. Immunol.
53
,
101532
https://doi.org/10.1016/j.smim.2021.101532
5
Mayer
,
D.A.
and
Fried
,
B
. (
2007
)
The role of helminth infections in carcinogenesis
.
Adv. Parasitol.
65
,
239
296
https://doi.org/10.1016/S0065-308X(07)65004-0
6
Natukunda
,
A.
,
Zirimenya
,
L.
,
Nassuuna
,
J.
,
Nkurunungi
,
G.
,
Cose
,
S.
,
Elliott
,
A.M.
et al.
(
2022
)
The effect of helminth infection on vaccine responses in humans and animal models: a systematic review and meta-analysis
.
Parasite Immunol.
44
, e12939 https://doi.org/10.1111/pim.12939
7
Acharya
,
S.
,
Da’dara
,
A.A.
and
Skelly
,
P.J
. (
2022
)
Schistosome immunomodulators
.
Plos Pathog.
17
, e1010064 https://doi.org/10.1371/journal.ppat.1010064
8
Bobardt
,
S.D.
,
Dillman
,
A.R.
and
Nair
,
M.G
. (
2020
)
The two faces of nematode infection: virulence and immunomodulatory molecules from nematode parasites of insects and mammals
.
Front. Microbiol.
11
, 577846 https://doi.org/10.3389/fmicb.2020.577846
9
Harnett
,
W
. (
2014
)
Secretory products of helminth parasites as immunomodulators
.
Mol. Biochem. Parasitol.
195
,
130
136
https://doi.org/10.1016/j.molbiopara.2014.03.007
10
Maizels
,
R.M.
,
Smits
,
H.H.
and
McSorley
,
H.J
. (
2018
)
Modulation of host immunity by helminths: the expanding repertoire of parasite effector molecules
.
Immunity
49
,
801
818
https://doi.org/10.1016/j.immuni.2018.10.016
11
Ryan
,
S.M.
,
Eichenberger
,
R.M.
,
Ruscher
,
R.
,
Giacomin
,
P.R.
and
Loukas
,
A
. (
2020
)
Harnessing helminth-driven immunoregulation in the search for novel therapeutic modalities
.
Plos Pathog.
16
, e1008508 https://doi.org/10.1371/journal.ppat.1008508
12
Lambrecht
,
B.N.
,
Hammad
,
H.
and
Fahy
,
J.V
. (
2019
)
The cytokines of asthma
.
Immunity
50
,
975
991
https://doi.org/10.1016/j.immuni.2019.03.018
13
Leonard
,
W.J.
,
Lin
,
J.X.
and
O’Shea
,
J.J
. (
2019
)
The γc family of cytokines: basic biology to therapeutic ramifications
.
Immunity
50
,
832
850
https://doi.org/10.1016/j.immuni.2019.03.028
14
Mantovani
,
A.
,
Dinarello
,
C.A.
,
Molgora
,
M.
and
Garlanda
,
C
. (
2019
)
Interleukin-1 and related cytokines in the regulation of inflammation and immunity
.
Immunity
50
,
778
795
https://doi.org/10.1016/j.immuni.2019.03.012
15
McGeachy
,
M.J.
,
Cua
,
D.J.
and
Gaffen
,
S.L
. (
2019
)
The IL-17 family of cytokines in health and disease
.
Immunity
50
,
892
906
https://doi.org/10.1016/j.immuni.2019.03.021
16
Massagué
,
J.
and
Sheppard
,
D
. (
2023
)
TGF-β signaling in health and disease
.
Cell
186
,
4007
4037
https://doi.org/10.1016/j.cell.2023.07.036
17
Richardson
,
L.
,
Wilcockson
,
S.G.
,
Guglielmi
,
L.
and
Hill
,
C.S
. (
2023
)
Context-dependent TGFβ family signalling in cell fate regulation
.
Nat. Rev. Mol. Cell Biol.
24
,
876
894
https://doi.org/10.1038/s41580-023-00638-3
18
Li
,
M.O.
,
Wan
,
Y.Y.
,
Sanjabi
,
S.
,
Robertson
,
A.K.
and
Flavell
,
R.A
. (
2006
)
Transforming growth factor-β regulation of immune responses
.
Annu. Rev. Immunol.
24
,
99
146
https://doi.org/10.1146/annurev.immunol.24.021605.090737
19
Kulkarni
,
A.B.
,
Thyagarajan
,
T.
and
Letterio
,
J.J
. (
2002
)
Function of cytokines within the TGF-β superfamily as determined from transgenic and gene knockout studies in mice
.
Curr. Mol. Med.
2
,
303
327
https://doi.org/10.2174/1566524024605699
20
Sanford
,
L.P.
,
Ormsby
,
I.
,
Gittenberger-de Groot
,
A.C.
,
Sariola
,
H.
,
Friedman
,
R.
,
Boivin
,
G.P.
et al.
(
1997
)
TGFβ2 knockout mice have multiple developmental defects that are non-overlapping with other TGFβ knockout phenotypes
.
Development
124
,
2659
2670
https://doi.org/10.1242/dev.124.13.2659
21
Kaartinen
,
V.
,
Voncken
,
J.W.
,
Shuler
,
C.
,
Warburton
,
D.
,
Bu
,
D.
,
Heisterkamp
,
N.
et al.
(
1995
)
Abnormal lung development and cleft palate in mice lacking TGF-β 3 indicates defects of epithelial-mesenchymal interaction
.
Nat. Genet.
11
,
415
421
https://doi.org/10.1038/ng1295-415
22
Ten Dijke
,
P
. (
2021
)
Therapeutic targeting of TGF-β in cancer: hacking a master switch of immune suppression
.
Clin. Sci. (Lond)
135
,
35
52
https://doi.org/10.1042/CS20201236
23
Tauriello
,
D.V.F.
,
Palomo-Ponce
,
S.
,
Stork
,
D.
,
Berenguer-Llergo
,
A.
,
Badia-Ramentol
,
J.
,
Iglesias
,
M.
et al.
(
2018
)
TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis
.
Nature
554
,
538
543
https://doi.org/10.1038/nature25492
24
Mariathasan
,
S.
,
Turley
,
S.J.
,
Nickles
,
D.
,
Castiglioni
,
A.
,
Yuen
,
K.
,
Wang
,
Y.
et al.
(
2018
)
TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells
.
Nature
554
,
544
548
https://doi.org/10.1038/nature25501
25
Maizels
,
R.M
. (
2021
)
The multi-faceted roles of TGF-β in regulation of immunity to infection
.
Adv. Immunol.
150
,
1
42
https://doi.org/10.1016/bs.ai.2021.05.001
26
Doetze
,
A.
,
Satoguina
,
J.
,
Burchard
,
G.
,
Rau
,
T.
,
Löliger
,
C.
,
Fleischer
,
B.
et al.
(
2000
)
Antigen-specific cellular hyporesponsiveness in a chronic human helminth infection is mediated by Th3/Tr1-type cytokines IL-10 and transforming growth factor-β but not by a Th1 to Th2 shift
.
Int. Immunol.
12
,
623
630
https://doi.org/10.1093/intimm/12.5.623
27
Araújo-Jorge
,
T.C.
,
Waghabi
,
M.C.
,
Soeiro
,
M. de N.C.
,
Keramidas
,
M.
,
Bailly
,
S.
and
Feige
,
J.J
. (
2008
)
Pivotal role for TGF-β in infectious heart disease: the case of Trypanosoma cruzi infection and consequent chagasic myocardiopathy
.
Cytokine Growth Factor Rev.
19
,
405
413
https://doi.org/10.1016/j.cytogfr.2008.08.002
28
Grainger
,
J.R.
,
Smith
,
K.A.
,
Hewitson
,
J.P.
,
McSorley
,
H.J.
,
Harcus
,
Y.
,
Filbey
,
K.J.
et al.
(
2010
)
Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-β pathway
.
J. Exp. Med.
207
,
2331
2341
https://doi.org/10.1084/jem.20101074
29
Worthington
,
J.J.
,
Klementowicz
,
J.E.
,
Rahman
,
S.
,
Czajkowska
,
B.I.
,
Smedley
,
C.
,
Waldmann
,
H.
et al.
(
2013
)
Loss of the TGFβ-activating integrin αvβ8 on dendritic cells protects mice from chronic intestinal parasitic infection via control of type 2 immunity
.
Plos Pathog.
9
, e1003675 https://doi.org/10.1371/journal.ppat.1003675
30
Smith
,
K.A.
,
Filbey
,
K.J.
,
Reynolds
,
L.A.
,
Hewitson
,
J.P.
,
Harcus
,
Y.
,
Boon
,
L.
et al.
(
2016
)
Low-level regulatory T-cell activity is essential for functional type-2 effector immunity to expel gastrointestinal helminths
.
Mucosal Immunol.
9
,
428
443
https://doi.org/10.1038/mi.2015.73
31
Hartmann
,
W.
,
Schramm
,
C.
and
Breloer
,
M
. (
2015
)
Litomosoides sigmodontis induces TGF-β receptor responsive, IL-10-producing T cells that suppress bystander T-cell proliferation in mice
.
Eur. J. Immunol.
45
,
2568
2581
https://doi.org/10.1002/eji.201545503
32
Hewitson
,
J.P.
,
Harcus
,
Y.
,
Murray
,
J.
,
van Agtmaal,
,
M.
,
Filbey
,
K.J.
,
Grainger
,
J.R.
et al.
(
2011
)
Proteomic analysis of secretory products from the model gastrointestinal nematode Heligmosomoides polygyrus reveals dominance of venom allergen-like (VAL) proteins
.
J. Proteomics
74
,
1573
1594
https://doi.org/10.1016/j.jprot.2011.06.002
33
Hewitson
,
J.P.
,
Ivens
,
A.C.
,
Harcus
,
Y.
,
Filbey
,
K.J.
,
McSorley
,
H.J.
,
Murray
,
J.
et al.
(
2013
)
Secretion of protective antigens by tissue-stage nematode larvae revealed by proteomic analysis and vaccination-induced sterile immunity
.
Plos Pathog.
9
, e1003492 https://doi.org/10.1371/journal.ppat.1003492
34
Johnston
,
C.J.C.
,
Smyth
,
D.J.
,
Kodali
,
R.B.
,
White
,
M.P.J.
,
Harcus
,
Y.
,
Filbey
,
K.J.
et al.
(
2017
)
A structurally distinct TGF-β mimic from an intestinal helminth parasite potently induces regulatory T cells
.
Nat. Commun.
8
,
1741
https://doi.org/10.1038/s41467-017-01886-6
35
Hinck
,
A.P.
,
Mueller
,
T.D.
and
Springer
,
T.A
. (
2016
)
Structural biology and evolution of the TGF-β family
.
Cold Spring Harb. Perspect. Biol.
8
, a022103 https://doi.org/10.1101/cshperspect.a022103
36
Mukundan
,
A.
,
Byeon
,
C.H.
,
Hinck
,
C.S.
,
Cunningham
,
K.
,
Campion
,
T.
,
Smyth
,
D.J.
et al.
(
2022
)
Convergent evolution of a parasite-encoded complement control protein-scaffold to mimic binding of mammalian TGF-β to its receptors, TβRI and TβRII
.
J. Biol. Chem.
298
, 101994 https://doi.org/10.1016/j.jbc.2022.101994
37
Liénart
,
S.
,
Merceron
,
R.
,
Vanderaa
,
C.
,
Lambert
,
F.
,
Colau
,
D.
,
Stockis
,
J.
et al.
(
2018
)
Structural basis of latent TGF-β1 presentation and activation by GARP on human regulatory T cells
.
Science
362
,
952
956
https://doi.org/10.1126/science.aau2909
38
Shi
,
M.
,
Zhu
,
J.
,
Wang
,
R.
,
Chen
,
X.
,
Mi
,
L.
,
Walz
,
T.
et al.
(
2011
)
Latent TGF-β structure and activation
.
Nature
474
,
343
349
https://doi.org/10.1038/nature10152
39
White
,
M.P.J.
,
Smyth
,
D.J.
,
Cook
,
L.
,
Ziegler
,
S.F.
,
Levings
,
M.K.
and
Maizels
,
R.M
. (
2021
)
The parasite cytokine mimic Hp-TGM potently replicates the regulatory effects of TGF-β on murine CD4+ T cells
.
Immunol. Cell Biol.
99
,
848
864
https://doi.org/10.1111/imcb.12479
40
Cook
,
L.
,
Reid
,
K.T.
,
Häkkinen
,
E.
,
de Bie,
,
B.
,
Tanaka
,
S.
,
Smyth
,
D.J.
et al.
(
2021
)
Induction of stable human FOXP3+ Tregs by a parasite-derived TGF-β mimic
.
Immunol. Cell Biol.
99
,
833
847
https://doi.org/10.1111/imcb.12475
41
Lothstein
,
K.E.
,
Chen
,
F.
,
Mishra
,
P.
,
Smyth
,
D.J.
,
Wu
,
W.
,
Lemenze
,
A.
et al.
(
2024
)
Helminth protein enhances wound healing by inhibiting fibrosis and promoting tissue regeneration
.
Life Sci. Alliance
7
, e202302249 https://doi.org/10.26508/lsa.202302249
42
Smyth
,
D.J.
,
White
,
M.P.J.
,
Johnston
,
C.J.C.
,
Donachie
,
A.M.
,
Campillo Poveda
,
M.
,
McSorley
,
H.J.
et al.
(
2023
)
Protection from T cell-dependent colitis by the helminth-derived immunomodulatory mimic of transforming growth factor-β, Hp-TGM
.
Discov. Immunol.
2
, kyad001 https://doi.org/10.1093/discim/kyad001
43
Chauché
,
C.
,
Rasid
,
O.
,
Donachie
,
A.M.
,
McManus
,
C.M.
,
Löser
,
S.
,
Campion
,
T.
et al.
(
2022
)
Suppression of airway allergic eosinophilia by Hp-TGM, a helminth mimic of TGF-β
.
Immunology
167
,
197
211
https://doi.org/10.1111/imm.13528
44
López-Casillas
,
F.
,
Wrana
,
J.L.
and
Massagué
,
J
. (
1993
)
Betaglycan presents ligand to the TGF-β signaling receptor
.
Cell
73
,
1435
1444
https://doi.org/10.1016/0092-8674(93)90368-z
45
Kanamori
,
M.
,
Nakatsukasa
,
H.
,
Okada
,
M.
,
Lu
,
Q.
and
Yoshimura
,
A
. (
2016
)
Induced regulatory T cells: their development, stability, and applications
.
Trends Immunol.
37
,
803
811
https://doi.org/10.1016/j.it.2016.08.012
46
Smyth
,
D.J.
,
Ren
,
B.
,
White
,
M.P.J.
,
McManus
,
C.
,
Webster
,
H.
,
Shek
,
V.
et al.
(
2021
)
Oral delivery of a functional algal-expressed TGF-β mimic halts colitis in a murine DSS model
.
J. Biotechnol.
340
,
1
12
https://doi.org/10.1016/j.jbiotec.2021.08.006
47
Redgrave
,
R.E.
,
Singh
,
E.
,
Tual-Chalot
,
S.
,
Park
,
C.
,
Hall
,
D.
,
Bennaceur
,
K.
et al.
(
2024
)
Exogenous transforming growth factor-β1 and its helminth-derived mimic attenuate the heart’s inflammatory ersponse to ischemic injury and reduce mature scar size
.
Am. J. Pathol.
194
,
562
573
https://doi.org/10.1016/j.ajpath.2023.09.014
48
Smyth
,
D.J.
,
Harcus
,
Y.
,
White
,
M.P.J.
,
Gregory
,
W.F.
,
Nahler
,
J.
,
Stephens
,
I.
et al.
(
2018
)
TGF-β mimic proteins form an extended gene family in the murine parasite Heligmosomoides polygyrus
.
Int. J. Parasitol.
48
,
379
385
https://doi.org/10.1016/j.ijpara.2017.12.004
49
Singh
,
S.P.
,
Smyth
,
D.J.
,
Cunningham
,
K.T.
,
Mukundan
,
A.
,
Byeon
,
C.H.
,
Hinck
,
C.S.
et al.
(
2025
)
The TGF-β mimic TGM4 achieves cell specificity through combinatorial surface co-receptor binding
.
EMBO Rep.
26
,
218
244
https://doi.org/10.1038/s44319-024-00323-2
50
White
,
S.E.
,
Schwartze
,
T.A.
,
Mukundan
,
A.
,
Schoenherr
,
C.
,
Singh
,
S.P.
,
van Dinther,
,
M.
et al.
(
2025
)
TGM6 is a helminth secretory product that mimics TGF-β binding to TGFBR2 to antagonize signaling in fibroblasts
.
Nat. Commun.
16
,
1847
https://doi.org/10.1038/s41467-025-56954-z
51
Maizels
,
R.M.
and
Newfeld
,
S.J
. (
2023
)
Convergent evolution in a murine intestinal parasite rapidly created the TGM family of molecular mimics to suppress the host immune response
.
Genome Biol. Evol.
15
, evad158 https://doi.org/10.1093/gbe/evad158
52
Maruszewska-Cheruiyot
,
M.
,
Szewczak
,
L.
,
Krawczak-Wójcik
,
K.
,
Głaczyńska
,
M.
and
Donskow-Łysoniewska
,
K
. (
2021
)
The production of excretory-secretory molecules from Heligmosomoides polygyrus bakeri fourth stage larvae varies between mixed and single sex cultures
.
Parasit. Vectors
14
,
106
https://doi.org/10.1186/s13071-021-04613-9
53
Groppe
,
J.
,
Hinck
,
C.S.
,
Samavarchi-Tehrani
,
P.
,
Zubieta
,
C.
,
Schuermann
,
J.P.
,
Taylor
,
A.B.
et al.
(
2008
)
Cooperative assembly of TGF-β superfamily signaling complexes is mediated by two disparate mechanisms and distinct modes of receptor binding
.
Mol. Cell
29
,
157
168
https://doi.org/10.1016/j.molcel.2007.11.039
54
van Dinther,
,
M.
,
Cunningham
,
K.T.
,
Singh
,
S.P.
,
White
,
M.P.J.
,
Campion
,
T.
,
Ciancia
,
C.
et al.
(
2023
)
CD44 acts as a coreceptor for cell-specific enhancement of signaling and regulatory T cell induction by TGM1, a parasite TGF-β mimic
.
Proc. Natl. Acad. Sci. U.S.A.
120
, e2302370120 https://doi.org/10.1073/pnas.2302370120
55
DeGrendele
,
H.C.
,
Estess
,
P.
,
Picker
,
L.J.
and
Siegelman
,
MH
. (
1996
)
CD44 and its ligand hyaluronate mediate rolling under physiologic flow: a novel lymphocyte-endothelial cell primary adhesion pathway
.
J. Exp. Med.
183
,
1119
1130
https://doi.org/10.1084/jem.183.3.1119
56
Ponta
,
H.
,
Sherman
,
L.
and
Herrlich
,
P.A
. (
2003
)
CD44: from adhesion molecules to signalling regulators
.
Nat. Rev. Mol. Cell Biol.
4
,
33
45
https://doi.org/10.1038/nrm1004
57
Budd
,
R.C.
,
Cerottini
,
J.C.
,
Horvath
,
C.
,
Bron
,
C.
,
Pedrazzini
,
T.
,
Howe
,
R.C.
et al.
(
1987
)
Distinction of virgin and memory T lymphocytes. stable acquisition of the Pgp-1 glycoprotein concomitant with antigenic stimulation
.
J. Immunol.
138
,
3120
3129
https://doi.org/10.4049/jimmunol.138.10.3120
58
Walter
,
R.J.
,
Sonnentag
,
S.J.
,
Munoz-Sagredo
,
L.
,
Merkel
,
M.
,
Richert
,
L.
,
Bunert
,
F.
et al.
(
2022
)
Wnt signaling is boosted during intestinal regeneration by a CD44-positive feedback loop
.
Cell Death Dis.
13
,
168
https://doi.org/10.1038/s41419-022-04607-0
59
Laiho
,
M.
,
Weis
,
M.B.
and
Massagué
,
J
. (
1990
)
Concomitant loss of transforming growth factor (TGF)-β receptor types I and II in TGF-β-resistant cell mutants implicates both receptor types in signal transduction
.
J. Biol. Chem.
265
,
18518
18524
https://doi.org/10.1016/S0021-9258(17)44782-X
60
Lin
,
H.Y.
,
Moustakas
,
A.
,
Knaus
,
P.
,
Wells
,
R.G.
,
Henis
,
Y.I.
and
Lodish
,
H.F
. (
1995
)
The soluble exoplasmic domain of the type II transforming growth factor (TGF)-β receptor. a heterogeneously glycosylated protein with high affinity and selectivity for TGF-β ligands
.
J. Biol. Chem.
270
,
2747
2754
https://doi.org/10.1074/jbc.270.6.2747
61
Stenvers
,
K.L.
,
Tursky
,
M.L.
,
Harder
,
K.W.
,
Kountouri
,
N.
,
Amatayakul-Chantler
,
S.
,
Grail
,
D.
et al.
(
2003
)
Heart and liver defects and reduced transforming growth factor β2 sensitivity in transforming growth factor β type III receptor-deficient embryos
.
Mol. Cell. Biol.
23
,
4371
4385
https://doi.org/10.1128/MCB.23.12.4371-4385.2003
62
Cheifetz
,
S.
,
Hernandez
,
H.
,
Laiho
,
M.
,
ten Dijke,
,
P.
,
Iwata
,
K.K.
and
Massagué
,
J
. (
1990
)
Distinct transforming growth factor-β (TGF-β) receptor subsets as determinants of cellular responsiveness to three TGF-β isoforms
.
J. Biol. Chem.
265
,
20533
20538
https://doi.org/10.1016/S0021-9258(17)30535-5
63
Wieteska
,
L.
,
Taylor
,
A.B.
,
Punch
,
E.
,
Coleman
,
J.A.
,
Conway
,
I.O.
,
Lin
,
Y.F.
et al.
(
2025
)
Structures of TGF-β with betaglycan and signaling receptors reveal mechanisms of complex assembly and signaling
.
Nat. Commun.
16
,
1778
https://doi.org/10.1038/s41467-025-56796-9
64
Herpin
,
A.
,
Lelong
,
C.
and
Favrel
,
P
. (
2004
)
Transforming growth factor-β-related proteins: an ancestral and widespread superfamily of cytokines in metazoans
.
Dev. Comp. Immunol.
28
,
461
485
https://doi.org/10.1016/j.dci.2003.09.007
65
Huminiecki
,
L.
,
Goldovsky
,
L.
,
Freilich
,
S.
,
Moustakas
,
A.
,
Ouzounis
,
C.
and
Heldin
,
CH
. (
2009
)
Emergence, development and diversification of the TGF-β signalling pathway within the animal kingdom
.
BMC Evol. Biol.
9
,
28
https://doi.org/10.1186/1471-2148-9-28
66
Gumienny
,
T.L.
and
Savage-Dunn
,
C
. (
2013
)
TGF-β signaling in C. elegans
.
WormBook
2013
,
1
34
https://doi.org/10.1895/wormbook.1.22.2
67
Ciccarelli
,
E.J.
,
Wing
,
Z.
,
Bendelstein
,
M.
,
Johal
,
R.K.
,
Singh
,
G.
,
Monas
,
A.
et al.
(
2024
)
TGF-β ligand cross-subfamily interactions in the response of Caenorhabditis elegans to a bacterial pathogen
.
Plos Genet.
20
, e1011324 https://doi.org/10.1371/journal.pgen.1011324
68
Estevez
,
M.
,
Attisano
,
L.
,
Wrana
,
J.L.
,
Albert
,
P.S.
,
Massagué
,
J.
and
Riddle
,
D.L
. (
1993
)
The daf-4 gene encodes a bone morphogenetic protein receptor controlling C. elegans dauer larva development
.
Nature
365
,
644
649
https://doi.org/10.1038/365644a0
69
Ren
,
P.
,
Lim
,
C.S.
,
Johnsen
,
R.J.
,
Albert
,
P.S.
,
Pilgrim
,
D.
and
Riddle
,
D.L
. (
1996
)
Control of C. elegans larval development by neuronal expression of a TGF-β homolog
.
Science
274
,
1389
1391
https://doi.org/10.1126/science.274.5291.1389
70
Gomez-Escobar
,
N.
,
van den Biggelaar,
,
A.
,
Maizels
,
R
. (
1997
)
A member of the TGF-β receptor gene family in the parasitic nematode Brugia pahangi
.
Gene
199
,
101
109
https://doi.org/10.1016/s0378-1119(97)00353-3
71
Gomez-Escobar
,
N.
,
Gregory
,
W.F.
and
Maizels
,
R.M
. (
2000
)
Identification of tgh-2, a filarial nematode homolog of Caenorhabditis elegans daf-7 and human transforming growth factor β, expressed in microfilarial and adult stages of Brugia malayi
.
Infect. Immun.
68
,
6402
6410
https://doi.org/10.1128/IAI.68.11.6402-6410.2000
72
Freitas
,
T.C.
and
Arasu
,
P
. (
2005
)
Cloning and characterisation of genes encoding two transforming growth factor-β-like ligands from the hookworm, Ancylostoma caninum
.
Int. J. Parasitol.
35
,
1477
1487
https://doi.org/10.1016/j.ijpara.2005.07.005
73
McSorley
,
H.J.
,
Grainger
,
J.R.
,
Harcus
,
Y.
,
Murray
,
J.
,
Nisbet
,
A.J.
,
Knox
,
D.P.
et al.
(
2010
)
daf-7-related TGF-β homologues from Trichostrongyloid nematodes show contrasting life-cycle expression patterns
.
Parasitology
137
,
159
171
https://doi.org/10.1017/S0031182009990321
74
He
,
L.
,
Gasser
,
R.B.
,
Li
,
T.
,
Di
,
W.
,
Li
,
F.
,
Zhang
,
H.
et al.
(
2019
)
A TGF-β type II receptor that associates with developmental transition in Haemonchus contortus in vitro
.
Plos Negl. Trop. Dis.
13
, e0007913 https://doi.org/10.1371/journal.pntd.0007913
75
He
,
L.
,
Liu
,
H.
,
Zhang
,
B.Y.
,
Li
,
F.F.
,
Di
,
W.D.
,
Wang
,
C.Q.
et al.
(
2020
)
A daf-7-related TGF-β ligand (Hc-tgh-2) shows important regulations on the development of Haemonchus contortus
.
Parasit. Vectors
13
,
326
https://doi.org/10.1186/s13071-020-04196-x
76
Freitas
,
T.C.
,
Jung
,
E.
and
Pearce
,
E.J
. (
2007
)
TGF-β signaling controls embryo development in the parasitic flatworm Schistosoma mansoni
.
Plos Pathog.
3
, e52 https://doi.org/10.1371/journal.ppat.0030052
77
Sulaiman
,
A.A.
,
Zolnierczyk
,
K.
,
Japa
,
O.
,
Owen
,
J.P.
,
Maddison
,
B.C.
,
Emes
,
R.D.
et al.
(
2016
)
A trematode parasite derived growth factor binds and exerts influences on host immune functions via host cytokine receptor complexes
.
Plos Pathog.
12
, e1005991 https://doi.org/10.1371/journal.ppat.1005991
78
Nono
,
J.K.
,
Lutz
,
M.B.
and
Brehm
,
KR
. (
2020
)
Expansion of host regulatory T cells by secreted products of the tapeworm Echinococcus multilocularis
.
Front. Immunol.
11
,
798
https://doi.org/10.3389/fimmu.2020.00798
79
Viney
,
M.E.
,
Thompson
,
F.J.
and
Crook
,
M
. (
2005
)
TGF-β and the evolution of nematode parasitism
.
Int. J. Parasitol.
35
,
1473
1475
https://doi.org/10.1016/j.ijpara.2005.07.006
80
Japa
,
O.
,
Hodgkinson
,
J.E.
,
Emes
,
R.D.
and
Flynn
,
RJ
. (
2015
)
TGF-β superfamily members from the helminth Fasciola hepatica show intrinsic effects on viability and development
.
Vet. Res.
46
,
29
https://doi.org/10.1186/s13567-015-0167-2
81
Musah-Eroje
,
M.
,
Hoyle
,
R.C.
,
Japa
,
O.
,
Hodgkinson
,
J.E.
,
Haig
,
D.M.
and
Flynn
,
R.J
. (
2021
)
A host-independent role for Fasciola hepatica transforming growth factor-like molecule in parasite development
.
Int. J. Parasitol.
51
,
481
492
https://doi.org/10.1016/j.ijpara.2020.11.005
82
Johnston
,
C.J.C.
,
Smyth
,
D.J.
,
Dresser
,
D.W.
and
Maizels
,
R.M
. (
2016
)
TGF-β in tolerance, development and regulation of immunity
.
Cell. Immunol.
299
,
14
22
https://doi.org/10.1016/j.cellimm.2015.10.006
83
Huber
,
S.
,
Stahl
,
F.R.
,
Schrader
,
J.
,
Lüth
,
S.
,
Presser
,
K.
,
Carambia
,
A.
et al.
(
2009
)
Activin a promotes the TGF-β-induced conversion of CD4⁺CD25⁻ T cells into Foxp3⁺ induced regulatory T cells
.
J. Immunol.
182
,
4633
4640
https://doi.org/10.4049/jimmunol.0803143
84
Osman
,
A.
,
Niles
,
E.G.
,
Verjovski-Almeida
,
S.
and
LoVerde
,
P.T
. (
2006
)
Schistosoma mansoni TGF-β receptor II: role in host ligand-induced regulation of a schistosome target gene
.
Plos Pathog
2
, e54 https://doi.org/10.1371/journal.ppat.0020054
85
Bancroft
,
A.J.
,
McKenzie
,
A.N.J.
and
Grencis
,
R.K
. (
1998
)
A critical role for IL-13 in resistance to intestinal nematode infection
.
J. Immunol.
160
,
3453
3461
https://doi.org/10.4049/jimmunol.160.7.3453
86
Bancroft
,
A.J.
,
Levy
,
C.W.
,
Jowitt
,
T.A.
,
Hayes
,
K.S.
,
Thompson
,
S.
,
Mckenzie
,
E.A.
et al.
(
2019
)
The major secreted protein of the whipworm parasite tethers to matrix and inhibits interleukin-13 function
.
Nat. Commun.
10
,
2344
https://doi.org/10.1038/s41467-019-09996-z
87
Foth
,
B.J.
,
Tsai
,
I.J.
,
Reid
,
A.J.
,
Bancroft
,
A.J.
,
Nichol
,
S.
,
Tracey
,
A.
et al.
(
2014
)
Whipworm genome and dual-species transcriptome analyses provide molecular insights into an intimate host-parasite interaction
.
Nat. Genet.
46
,
693
700
https://doi.org/10.1038/ng.3010
88
McSorley
,
H.J.
,
Blair
,
N.F.
,
Smith
,
K.A.
,
McKenzie
,
A.N.J.
and
Maizels
,
R.M
. (
2014
)
Blockade of IL-33 release and suppression of type 2 innate lymphoid cell responses by helminth secreted products in airway allergy
.
Mucosal Immunol.
7
,
1068
1078
https://doi.org/10.1038/mi.2013.123
89
Coakley
,
G.
,
McCaskill
,
J.L.
,
Borger
,
J.G.
,
Simbari
,
F.
,
Robertson
,
E.
,
Millar
,
M.
et al.
(
2017
)
Extracellular vesicles from a helminth parasite suppress macrophage activation and constitute an effective vaccine for protective immunity
.
Cell Rep.
19
,
1545
1557
https://doi.org/10.1016/j.celrep.2017.05.001
90
Zaiss
,
M.M.
,
Maslowski
,
K.M.
,
Mosconi
,
I.
,
Guenat
,
N.
,
Marsland
,
B.J.
and
Harris
,
N.L
. (
2013
)
IL-1β suppresses innate IL-25 and IL-33 production and maintains helminth chronicity
.
Plos Pathog.
9
, e1003531 https://doi.org/10.1371/journal.ppat.1003531
91
Jamwal
,
A.
,
Colomb
,
F.
,
McSorley
,
H.J.
and
Higgins
,
M.K
. (
2024
)
Structural basis for IL-33 recognition and its antagonism by the helminth effector protein HpARI2
.
Nat. Commun.
15
,
5226
https://doi.org/10.1038/s41467-024-49550-0
92
Osbourn
,
M.
,
Soares
,
D.C.
,
Vacca
,
F.
,
Cohen
,
E.S.
,
Scott
,
I.C.
,
Gregory
,
W.F.
et al.
(
2017
)
HpARI protein secreted by a helminth parasite suppresses interleukin-33
.
Immunity
47
,
739
751
https://doi.org/10.1016/j.immuni.2017.09.015
93
Colomb
,
F.
,
Ogunkanbi
,
A.
,
Jamwal
,
A.
,
Dong
,
B.
,
Maizels
,
R.M.
,
Finney
,
C.A.M.
et al.
(
2024
)
IL-33-binding HpARI family homologues with divergent effects in suppressing or enhancing type 2 immune responses
.
Infect. Immun.
92
, e00395-23 https://doi.org/10.1128/iai.00395-23
94
Colomb
,
F.
,
Jamwal
,
A.
,
Ogunkanbi
,
A.
,
Frangova
,
T.
,
Savage
,
A.R.
,
Kelly
,
S.
et al.
(
2024
)
Heparan sulphate binding controls in vivo half-life of the HpARI protein family
.
Elife
13
, RP99000 https://doi.org/10.7554/eLife.99000
95
Vacca
,
F.
,
Chauché
,
C.
,
Jamwal
,
A.
,
Hinchy
,
E.C.
,
Heieis
,
G.
,
Webster
,
H.
et al.
(
2020
)
A helminth-derived suppressor of ST2 blocks allergic responses
.
Elife
9
, e54017 https://doi.org/10.7554/eLife.54017
96
Colomb
,
F.
and
McSorley
,
H.J
. (
2025
)
Protein families secreted by nematodes to modulate host immunity
.
Curr. Opin. Microbiol.
84
, 102582 https://doi.org/10.1016/j.mib.2025.102582
97
Varyani
,
F.
,
Löser
,
S.
,
Filbey
,
K.J.
,
Harcus
,
Y.
,
Drurey
,
C.
,
Poveda
,
M.C.
et al.
(
2022
)
The IL-25-dependent tuft cell circuit driven by intestinal helminths requires macrophage migration inhibitory factor (MIF)
.
Mucosal Immunol.
15
,
1243
1256
https://doi.org/10.1038/s41385-022-00496-w
98
Filbey
,
K.J.
,
Varyani
,
F.
,
Harcus
,
Y.
,
Hewitson
,
J.P.
,
Smyth
,
D.J.
,
McSorley
,
H.J.
et al.
(
2019
)
Macrophage migration inhibitory factor (MIF) is essential for type 2 effector cell immunity to an intestinal helminth parasite
.
Front. Immunol.
10
, 2375 https://doi.org/10.3389/fimmu.2019.02375
99
Sparkes
,
A.
,
De Baetselier
,
P.
,
Roelants
,
K.
,
De Trez
,
C.
,
Magez
,
S.
,
Van Ginderachter
,
J.A.
et al.
(
2017
)
The non-mammalian MIF superfamily
.
Immunobiology
222
,
473
482
https://doi.org/10.1016/j.imbio.2016.10.006
100
Prieto-Lafuente
,
L.
,
Gregory
,
W.F.
,
Allen
,
J.E.
and
Maizels
,
R.M
. (
2009
)
MIF homologues from a filarial nematode parasite synergize with IL-4 to induce alternative activation of host macrophages
.
J. Leukoc. Biol.
85
,
844
854
https://doi.org/10.1189/jlb.0808459
101
Younis
,
A.E.
,
Soblik
,
H.
,
Ajonina-Ekoti
,
I.
,
Erttmann
,
K.D.
,
Luersen
,
K.
,
Liebau
,
E.
et al.
(
2012
)
Characterization of a secreted macrophage migration inhibitory factor homologue of the parasitic nematode Strongyloides acting at the parasite–host cell interface
.
Microbes Infect.
14
,
279
289
https://doi.org/10.1016/j.micinf.2011.09.006
102
Park
,
S.K.
,
Cho
,
M.K.
,
Park
,
H.K.
,
Lee
,
K.H.
,
Lee
,
S.J.
,
Choi
,
S.H.
et al.
(
2009
)
Macrophage migration inhibitory factor homologs of anisakis simplex suppress Th2 response in allergic airway inflammation model via CD4⁺CD25⁺Foxp3⁺ T cell recruitment
.
J. Immunol.
182
,
6907
6914
https://doi.org/10.4049/jimmunol.0803533
103
Fitzsimmons
,
C.M.
,
Schramm
,
G.
,
Jones
,
F.M.
,
Chalmers
,
I.W.
,
Hoffmann
,
K.F.
,
Grevelding
,
C.G.
et al.
(
2005
)
Molecular characterization of omega-1: a hepatotoxic ribonuclease from Schistosoma mansoni eggs
.
Mol. Biochem. Parasitol.
144
,
123
127
https://doi.org/10.1016/j.molbiopara.2005.08.003
104
Everts
,
B.
,
Perona-Wright
,
G.
,
Smits
,
H.H.
,
Hokke
,
C.H.
,
van der Ham,
,
A.J.
,
Fitzsimmons
,
C.M.
et al.
(
2009
)
Omega-1, a glycoprotein secreted by Schistosoma mansoni eggs, drives Th2 responses
.
J. Exp. Med.
206
,
1673
1680
https://doi.org/10.1084/jem.20082460
105
Steinfelder
,
S.
,
Andersen
,
J.F.
,
Cannons
,
J.L.
,
Feng
,
C.G.
,
Joshi
,
M.
,
Dwyer
,
D.
et al.
(
2009
)
The major component in schistosome eggs responsible for conditioning dendritic cells for Th2 polarization is a T2 ribonuclease (omega-1)
.
J. Exp. Med.
206
,
1681
1690
https://doi.org/10.1084/jem.20082462
106
Everts
,
B.
,
Hussaarts
,
L.
,
Driessen
,
N.N.
,
Meevissen
,
M.H.J.
,
Schramm
,
G.
,
van der Ham,
,
A.J.
et al.
(
2012
)
Schistosome-derived omega-1 drives Th2 polarization by suppressing protein synthesis following internalization by the mannose receptor
.
J. Exp. Med.
209
,
1753
1767
https://doi.org/10.1084/jem.20111381
107
Ferguson
,
B.J.
,
Newland
,
S.A.
,
Gibbs
,
S.E.
,
Tourlomousis
,
P.
,
Fernandes dos Santos
,
P.
,
Patel
,
M.N.
et al.
(
2015
)
The Schistosoma mansoni T2 ribonuclease omega-1 modulates inflammasome-dependent IL-1β secretion in macrophages
.
Int. J. Parasitol.
45
,
809
813
https://doi.org/10.1016/j.ijpara.2015.08.005
108
Patente
,
T.A.
,
Gasan
,
T.A.
,
Scheenstra
,
M.
,
Ozir-Fazalalikhan
,
A.
,
Obieglo
,
K.
,
Schetters
,
S.
et al.
(
2024
)
S. mansoni -derived omega-1 prevents OVA-specific allergic airway inflammation via hampering of cDC2 migration
.
Plos Pathog.
20
, e1012457 https://doi.org/10.1371/journal.ppat.1012457
109
Zawistowska-Deniziak
,
A.
,
Lambooij
,
J.M.
,
Kalinowska
,
A.
,
Patente
,
T.A.
,
Łapiński
,
M.
,
van der Zande,
,
H.J.P.
et al.
(
2022
)
Fasciola hepatica fatty acid binding protein 1 modulates T cell polarization by promoting dendritic cell thrombospondin-1 secretion without affecting metabolic homeostasis in obese mice
.
Front. Immunol.
13
, 884663 https://doi.org/10.3389/fimmu.2022.884663
110
Pineda
,
M.A.
,
Lumb
,
F.
,
Harnett
,
M.M.
and
Harnett
,
W
. (
2014
)
ES-62, a therapeutic anti-inflammatory agent evolved by the filarial nematode Acanthocheilonema viteae
.
Mol. Biochem. Parasitol.
194
,
1
8
https://doi.org/10.1016/j.molbiopara.2014.03.003
111
Goodridge
,
H.S.
,
Marshall
,
F.A.
,
Else
,
K.J.
,
Houston
,
K.M.
,
Egan
,
C.
,
Al-Riyami
,
L.
et al.
(
2005
)
Immunomodulation via novel use of TLR4 by the filarial nematode phosphorylcholine-containing secreted product, ES-62
.
J. Immunol.
174
,
284
293
https://doi.org/10.4049/jimmunol.174.1.284
112
Haeberlein
,
S.
,
Obieglo
,
K.
,
Ozir-Fazalalikhan
,
A.
,
Chayé
,
M.A.M.
,
Veninga
,
H.
van der Vlugt,
,
L.E.P.M.
et al.
(
2017
)
Schistosome egg antigens, including the glycoprotein IPSE/alpha-1, trigger the development of regulatory B cells
.
Plos Pathog.
13
, e1006539 https://doi.org/10.1371/journal.ppat.1006539
113
Chayé
,
M.A.M.
,
Gasan
,
T.A.
,
Ozir-Fazalalikhan
,
A.
,
Scheenstra
,
M.R.
,
Zawistowska-Deniziak
,
A.
,
van Hengel,
,
O.R.J.
et al.
(
2023
)
Schistosoma mansoni egg-derived thioredoxin and Sm14 drive the development of IL-10 producing regulatory B cells
.
Plos Negl. Trop. Dis.
17
, e0011344 https://doi.org/10.1371/journal.pntd.0011344
114
Hams
,
E.
,
Bermingham
,
R.
,
Wurlod
,
F.A.
,
Hogan
,
A.E.
,
O’Shea
,
D.
,
Preston
,
R.J.
et al.
(
2016
)
The helminth T2 RNase ω1 promotes metabolic homeostasis in an IL-33- and group 2 innate lymphoid cell-dependent mechanism
.
FASEB J.
30
,
824
835
https://doi.org/10.1096/fj.15-277822
115
van der Zande,
,
H.J.P.
,
Gonzalez
,
M.A.
,
de Ruiter,
,
K.
,
Wilbers
,
R.H.P.
,
García-Tardón
,
N.
,
van Huizen,
,
M.
et al.
(
2021
)
The helminth glycoprotein omega-1 improves metabolic homeostasis in obese mice through type 2 immunity-independent inhibition of food intake
.
FASEB J.
35
, e21331 https://doi.org/10.1096/fj.202001973R
116
Schnoeller
,
C.
,
Rausch
,
S.
,
Pillai
,
S.
,
Avagyan
,
A.
,
Wittig
,
B.M.
,
Loddenkemper
,
C.
et al.
(
2008
)
A helminth immunomodulator reduces allergic and inflammatory responses by induction of IL-10-producing macrophages
.
J. Immunol.
180
,
4265
4272
https://doi.org/10.4049/jimmunol.180.6.4265
117
Ji
,
P.
,
Hu
,
H.
,
Yang
,
X.
,
Wei
,
X.
,
Zhu
,
C.
,
Liu
,
J.
et al.
(
2015
)
AcCystatin, an immunoregulatory molecule from Angiostrongylus cantonensis, ameliorates the asthmatic response in an aluminium hydroxide/ovalbumin-induced rat model of asthma
.
Parasitol. Res.
114
,
613
624
https://doi.org/10.1007/s00436-014-4223-z
118
Coronado
,
S.
,
Zakzuk
,
J.
,
Regino
,
R.
,
Ahumada
,
V.
,
Benedetti
,
I.
,
Angelina
,
A.
et al.
(
2019
)
Ascaris lumbricoides cystatin prevents development of allergic airway inflammation in a mouse model
.
Front. Immunol.
10
, 2280 https://doi.org/10.3389/fimmu.2019.02280
119
Acevedo
,
N.
,
Lozano
,
A.
,
Zakzuk
,
J.
,
Llinás-Caballero
,
K.
,
Brodin
,
D.
,
Nejsum
,
P.
et al.
(
2024
)
Cystatin from the helminth Ascaris lumbricoides upregulates mevalonate and cholesterol biosynthesis pathways and immunomodulatory genes in human monocyte-derived dendritic cells
.
Front. Immunol.
15
, 1328401 https://doi.org/10.3389/fimmu.2024.1328401
120
Quinteros
,
S.L.
,
von Krusenstiern,
,
E.
,
Snyder
,
N.W.
,
Tanaka
,
A.
,
O’Brien
,
B.
and
Donnelly
,
S
. (
2023
)
The helminth derived peptide FhHDM-1 redirects macrophage metabolism towards glutaminolysis to regulate the pro-inflammatory response
.
Front. Immunol.
14
, 1018076 https://doi.org/10.3389/fimmu.2023.1018076
121
Bohnacker
,
S.
,
Henkel
,
F.D.R.
,
Hartung
,
F.
,
Geerlof
,
A.
,
Riemer
,
S.
,
Prodjinotho
,
U.F.
et al.
(
2024
)
A helminth enzyme subverts macrophage-mediated immunity by epigenetic targeting of prostaglandin synthesis
.
Sci. Immunol.
9
, eadl1467 https://doi.org/10.1126/sciimmunol.adl1467
122
Martínez-Reyes
,
I.
and
Chandel
,
N.S
. (
2020
)
Mitochondrial TCA cycle metabolites control physiology and disease
.
Nat. Commun.
11
,
102
https://doi.org/10.1038/s41467-019-13668-3
123
Yu
,
X.
,
Orr
,
C.M.
,
Chan
,
H.T.C.
,
James
,
S.
,
Penfold
,
C.A.
,
Kim
,
J.
et al.
(
2023
)
Reducing affinity as a strategy to boost immunomodulatory antibody agonism
.
Nature
614
,
539
547
https://doi.org/10.1038/s41586-022-05673-2
124
Li
,
H.
,
Wang
,
J.
,
Kuan
,
T.A.
,
Tang
,
B.
,
Feng
,
L.
,
Wang
,
J.
et al.
(
2023
)
Pathogen protein modularity enables elaborate mimicry of a host phosphatase
.
Cell
186
,
3196
3207
https://doi.org/10.1016/j.cell.2023.05.049
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).