Abstract
Background
The detection of pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) by multimeric protein complexes, known as inflammasomes, triggers an inflammatory response, which is a critical component of the innate immune system. This inflammatory response plays a pivotal role in host resistance against parasitic infections, presenting a significant global health challenge.
Methods
We systematically searched for relevant articles from the Pubmed and the Web of Science database to summarize current insights into how inflammasomes function in preventing infections caused by the apicomplexan parasites Toxoplasma and Plasmodium.
Results
In vivo and in vitro studies have extensively explored inflammasomes such as the absent in melanoma 2 (AIM2), NLR family pyrin-containing protein 1 (NLRP1), NLRP3, and NLRP12 inflammasomes, alongside noncanonical inflammasomes, with particular emphasis on the NLRP1 and the NLRP3 inflammasome during Toxoplasma gondii infection or the AIM2 and the NLRP3 inflammasome at various stages of Plasmodium infection. Toxoplasma gondii interacts with inflammasomes to activate or inhibit immune responses.
Conclusions
Inflammasomes control parasite burden and parasite-induced cell death, contribute to immune recognition and inflammatory responses and thus influence apicomplexan parasite-associated pathogenesis and the severity of clinical outcomes. Hence, inflammasomes play crucial roles in the progression and outcomes of toxoplasmosis and malaria. A comprehensive understanding of how parasitic infections modulate inflammasome activity enhances insight into host immune responses against parasites.
Graphical Abstract
Keywords: T. gondii, Plasmodium, Inflammasomes, Infection
Background
Inflammasomes, which are integral to the innate immune system, are indispensable for defending the host against pathogens and regulating the autoinflammatory response. Excessive inflammation can result in cell and tissue damage, whereas an insufficient inflammatory response may enable pathogens to persist in the host.
Inflammasomes represent a diverse group of cytoplasmic multiprotein complexes that primarily consist of a sensor protein, an adaptor protein and an inflammatory caspase. These sensor proteins, which function as pattern recognition receptors (PRRs), identify pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), initiating the assembly of inflammasomes. Common sensor proteins include Nucleotide binding oligomerization domain-like receptor (NLR) family proteins (such as NLRP1, NLRP3 and NLRC4), PYRIN and AIM2. Canonical inflammasomes (NLRP1, NLRP3, NLRC4, AIM2) activate caspase-1, leading to the release of the proinflammatory cytokines interleukin-1 beta (IL-1β) and IL-18 and the cleavage of gasdermin D (GSDMD) into its N-terminus (GSDMD-NT), triggering inflammation or cell death. The noncanonical inflammasomes caspase-11 (in mice) and caspase-4/5 (in humans) directly bind lipopolysaccharide (LPS) from Gram-negative bacteria, triggering caspase oligomerization and activation, which is independent of caspase-1 activation. Caspase-4/5/11 cannot directly process pro-IL-1β and pro-IL-18 into its mature fragment, but it can induce the secretion of IL-1β and IL-18 via NLRP3 inflammasome activation or cellular pyroptosis. NLR family proteins typically feature an leucine-rich repeat (LRR) domain that binds ligands, a central nucleotide-binding and oligomerization domain (NACHT) and an N-terminal caspase-activating and recruitment domain (CARD) or pyrin domain (PYD). Procaspase-1 is recruited to inflammasomes through its CARD domain by apoptosis-associated speck-like protein containing a CARD (ASC). The activation of the NLRP3 inflammasome involves two main phases: priming and activation. Priming involves PAMPs or DAMPs such as LPS, which activate nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling and induce the transcription of the NLRP3, IL-18 and IL-1β genes, accompanied by posttranslational modifications (ubiquitination, deubiquitination, phosphorylation and dephosphorylation of NLRP3). Activation signals (e.g. ATP, nigericin, free fatty acids, urate crystals and cholesterol crystals) trigger cellular responses, including potassium efflux, calcium mobilization, mitochondrial dysfunction, reactive oxygen species (ROS) production and lysosomal rupture.
NLRP3 inflammasome components are expressed across a spectrum of cell types, including monocytes, neutrophils, macrophages, dendritic cells, oligodendrocytes, astrocytes, endothelial cells and retinal pigmented epithelial cells. They are closely associated with the pathogenesis of various diseases, such as viral, fungal and bacterial infections; atherosclerosis; Alzheimer's disease; cancer; and nonalcoholic fatty liver disease [1]. These findings suggest that the NLRP3 inflammasome is an increasingly important target for treating inflammatory conditions. Recent studies highlight the crucial role of inflammasomes in the process of antiparasitic infection, as they participate in the inflammatory response against antiprotozoal infections and potentially aid in parasite clearance and reducing parasite loads. In toxoplasmosis and malaria, inflammasomes are primed and activated, responding to parasite components that act as PAMPs recognized by host PRRs, thereby regulating inflammasome activation.
This review briefly summarizes recent developments in harnessing inflammasomes against Toxoplasma and Plasmodium infections.
Toxoplasma
The obligate intracellular protozoan parasite Toxoplasma gondii infects virtually all nucleated cell types in warm-blooded animals, including humans, leading to toxoplasmosis. This zoonotic disease typically spreads through the ingestion of oocysts or tissue cysts (bradyzoites) in contaminated food or water. Global estimates indicate that approximately 30–50% of the world's population is infected with T. gondii, with seroprevalence rates varying widely (from < 10% to > 70%) across different regions [2, 3]. Despite their high prevalence, the majority of infected individuals either show no clinical symptoms or experience only mild discomfort. In these cases, the immune system controls the replication of T. gondii tachyzoites, while some tachyzoites may transform into slow-growing and metabolically inactive bradyzoites, primarily found in organs such as muscle and brain tissues, establishing chronic infections. However, in individuals with a suppressed or compromised immune system, bradyzoites can convert back into active tachyzoites, posing significant health risks [4]. Moreover, T. gondii infection in pregnant women can lead to miscarriage or congenital toxoplasmosis in newborns, impacting public health [5].
Toxoplasma gondii infection hinges on a delicate balance between the host immune response and parasite immune evasion strategies, enabling survival for both parties. Monocytes and macrophages, pivotal components of the innate immune system, are swiftly recruited to sites of T. gondii infection and play crucial roles in defending against Toxoplasma. Monocytes contribute by regulating parasite levels through the secretion of inflammatory cytokines, phagocytosis and antimicrobial effector mechanisms [6]. Moreover, macrophages curb the growth and reproduction of T. gondii, although the parasite can evade phagocytosis and invade macrophages by preventing the recruitment of immunity-related GTPases to the parasitophorous vacuole membrane [7].
Recent studies have revealed that T. gondii, an intracellular pathogen, has the ability to regulate inflammasome activation in a bidirectional manner. Upon invading immune cells, T. gondii can either induce inflammasome activation or inhibit inflammasome activation as part of its immune evasion strategy. The parasite manipulates human neutrophils to evade innate immunity, where infected neutrophils suppress LPS-induced IL-1β and NLRP3 transcription, reduce the expression of pro-IL-1β and mature IL-1β and hinder NLRP3 inflammasome activation by disrupting NF-κB signaling. These mechanisms facilitate parasite propagation [8]. Consequently, inflammasomes play a limited role in host defense against T. gondii. In the following sections, we summarize recent advancements in understanding the role of inflammasomes during T. gondii infection (Fig. 1).
Fig. 1.
Inflammasomes mediate the immune response in host cells infected with Toxoplasma gondii. Toxoplasma gondii is involved primarily in NLRP1 and NLRP3 inflammasome activation in human macrophages, monocytes, small intestinal epithelial cells and mouse macrophages. TgGRA7, TgGRA15, profilin and TgROP7 are associated with NLRP1 and NLRP3 inflammasome activation. The P2X7 receptor and potassium efflux reportedly activate the NLRP3 inflammasome. Toxoplasma gondii activates a Syk-CARD9-NF-κB signaling axis, leading to NLRP3 inflammasome activation in primary human monocytes [9]. Toxoplasma gondii-induced NLRP3 inflammasome activation in human small intestinal epithelial cells is strongly correlated with the phosphorylation of p38 MAPK [10]. In addition, the NLRP1 inflammasome is activated in T. gondii-infected rat macrophages. TgGRA35, TgGRA42 and TgGRA43 are associated with NLRP1 inflammasome activation [11]. CARD, Caspase-activating and recruitment domain; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; NLRP, NLR family pyrin-containing protein TgGRA; T. gondii dense granule protein; TgROP7, T. gondii Rhoptry protein 7; see also Abbreviation list
The NLRP1, NLRP3 and AIM2 inflammasomes are actively involved in the immune response against T. gondii infection. The activation of the NLRP1 and the NLRP3 inflammasome contributes significantly to host defense by controlling Toxoplasma replication and limiting parasite proliferation. These inflammasomes are critical for generating IL-1β and IL-18, which are essential cytokines that play a key role in combating Toxoplasma in both in vitro and in vivo experimental models [12-14]. Studies using knockout mice lacking NLRP3, caspase-1/11, the IL-1 receptor or ASC have demonstrated reduced IL-18 levels, increased parasite growth, and increased mortality rates [13]. IL-18 cytokine production is pivotal in restraining parasite proliferation and enhancing survival in mice deficient in IL-18 or its receptor [13]. Notably, T. gondii infection induces both the activation and inhibition of caspases, influencing various outcomes in the responses of murine and human cells to the parasite [15].
NLRP1 inflammasome
The Lewis rat strain exhibits unexpected resistance to Toxoplasma infection, indicating that establishing effective infection through oral or intraperitoneal routes is difficult. This resistance trait is dominantly inherited and linked to bone marrow-derived cells (BMDMs) [14, 16]. Investigations have pinpointed the Toxo1 locus on rat chromosome 10, which spans an 891-kb region that includes the NLRP1 gene and governs Lewis rats' resistance to T. gondii [16]. Variations in the NLRP1 gene among rat strains determine differing susceptibilities to T. gondii [14, 16]. Silencing NLRP1 in Lewis rat macrophages increases parasite proliferation and provides protection against cell death, whereas overexpression of the NLRP1 variant in BMDMs from susceptible Fischer CDF rats sensitized these cells [14]. Additionally, the NLRP1b allele has been found to enhance the inflammasome response in B6 BMDMs to Toxoplasma, independent of the lethal factor proteolysis site [12]. The NLRP1a/Caspase-1 pathway is crucial for mediating parasite-induced macrophage death [16]. Consequently, Toxoplasma infection induces NLRP1 inflammasome-dependent release of the proinflammatory cytokine IL-1β in Lewis rat macrophages, inhibiting parasite proliferation [12, 14, 16]. This process triggers typical cellular pyroptosis, facilitating the clearance of intracellular parasites and protecting rats from toxoplasmosis [12, 14].
The dense granule proteins TgGRA35, TgGRA42 and TgGRA43, which are localized in the T. gondii parasitophorous vacuole, play critical roles in activating the NLRP1 inflammasome and promoting IL-1β secretion, resulting in pyroptosis of Lewis rat BMDMs and inhibition of parasite proliferation. Infection of macrophages with TgGRA35, TgGRA42 or TgGRA43 knockout parasites significantly reduces cell death rates and increases parasite replication. Additionally, TgGRA42 and TgGRA43 facilitate the proper localization of other TgGRAs, including TgGRA35, to the parasitophorous vacuole membrane [11]. TgGRA35 interacts with the host E3 ubiquitin ligase ITCH, leading to NLRP1 inflammasome activation. Inhibition of proteasome activity or knockout of ITCH prevents Toxoplasma-induced pyroptosis in Lewis rat macrophages. Interestingly, the TgGRA35-ITCH interaction contributes to restricting Toxoplasma growth in human fibroblasts stimulated with interferon gamma (IFN-γ) [17].
The NLRP1 inflammasome enhances the expression of proinflammatory cytokines such as IL-1β, IL-18 and IL-12. Silencing NLRP1 accelerates host cell death and leads to cell disintegration, which slows the progression of T. gondii infection in human monocytic cells [18]. Mice deficient in NLRP1 exhibit increased parasite burdens and higher mortality rates during T. gondii infection [13], highlighting the pivotal role of the NLRP1 inflammasome in modulating the pathogenesis of T. gondii infection.
NLRP3 inflammasome
The NLRP3 inflammasome plays a crucial role in clearing T. gondii and protecting the host. The activation of the NLRP3 inflammasome in response to Toxoplasma infection varies among different host cell types. In murine BMDMs, Toxoplasma induces NLRP3 inflammasome activation, leading to the rapid production and secretion of IL-1β [13]. However, primary human macrophages infected with T. gondii do not produce IL-1β, possibly because of the downregulation of NLRP3 during monocyte-to-macrophage differentiation and the lack of induction during infection [19]. Toxoplasma triggers the Syk-CARD9/MALT-1-NF-κB signaling pathway and activates the NLRP3-caspase-1 inflammasome in primary human monocytes, resulting in IL-1β release in a manner independent of cell death and GSDMD [9]. Additionally, T. gondii-induced NLRP3 inflammasome activation in human small intestinal epithelial cells is strongly correlated with p38 MAPK phosphorylation [10].
The activation of the purinergic receptor P2X7 is essential for regulating immune responses against T. gondii, reducing parasite load and promoting parasite clearance through canonical NLRP3 inflammasome activation in infected cells [20–23]. In human macrophages infected with T. gondii, P2X7 receptor activation decreases the parasite burden by inducing apoptosis in host cells [23]. Deficiency in the P2X7 receptor compromises the ability of both human and mouse macrophages to eliminate T. gondii. P2X7 receptor-mediated parasite death occurs alongside host cell apoptosis and operates independently of NO production [20]. ATP-induced activation of P2X7 inhibits T. gondii growth in macrophages by promoting NADPH oxidase-dependent ROS production and activating the NLRP3 inflammasome, which triggers IL-1β production through caspase-1 activity and mitochondrial ROS generation [21]. The P2X7 receptor plays a critical role in controlling the parasite load and mitigating tissue damage during virulent T. gondii infection [22]. However, in human small intestinal epithelial cells infected with T. gondii, P2X7 receptor-mediated NLRP3-dependent IL-1β secretion is crucial for regulating parasite replication. Knockdown of NLRP3, cleaved caspase-1, ASC or the P2X7 receptor significantly decreases T. gondii-induced IL-1β production and markedly increases parasite replication [24]. The P2X7 receptor regulates the upregulation of NLRP3 and IL-1β and the downregulation of nitric oxide (NO) following ileitis in T. gondii-infected mice. Compared with wild-type mice, the absence of the P2X7 receptor during T. gondii infection results in imbalanced ileal inflammatory responses, leading to increased intestinal parasite burdens, extensive inflammation and increased intestinal contractility. Also, compared with serum levels of IL-6, IFN-γ, tissue caspase-1 and NLRP3 in wild-type mice, T. gondii infection in P2X7-deficient mice elevates the serum levels of IL-6, IFN-γ and tissue caspase-1, but not NLRP3, [25]. Therefore, the P2X7 receptor, an activator of the NLRP3 inflammasome, plays a crucial role in determining susceptibility to congenital toxoplasmosis and ileitis [25–27].
In addition to ROS and the P2X7 receptor, potassium efflux represents a critical step for NLRP3 inflammasome activation in human monocytes during T. gondii infection [19]. The lethal type II strains of T. gondii, PRU tachyzoites, RNA and soluble tachyzoite antigen predominantly induce NLRP3 inflammasome activation [28]. Moreover, several immunomodulatory molecules produced by T. gondii regulate the activation of the NLRP3 inflammasome to control parasitic growth (Table 1).
Table 1.
Summary of the role of Toxoplasma gondii molecules in modulating NLRP3 inflammasome activation during parasitic infection
| Molecule | Description | Cells/animals | Function |
|---|---|---|---|
| Profilin [29] | Induces NLRP3 activation and IL-1β secretion | THP-1 cells | - |
| TgROP7 [30] | Interacts with the NACHT domain of NLRP3, leading to an increase in NF-κB and continuous secretion of IL-1β through the IL-1β/NF-κB/ NLRP3 feedback loop | Differentiated THP-1 cells | - |
| TgGRA9 [31] | Inhibits the formation of the NLRP3 inflammasome by preventing the binding of ASC to NLRP3 in mitochondria, and regulates NLRP3 inflammasome activation | - | Enhances its anti-inflammatory and bactericidal effects through M2 polarization and exhibits anti-septic properties |
| TgGRA15 [32,33,34] | Induces NLRP3 inflammasome activation or activates NF-κB pathway, resulting in IL-1β secretion | Monocytes | Induces proinflammatory responses, stimulates the host innate immune response |
| TgGRA7 [35,36] | Activates the MyD88-dependent NF-κB pathway, phosphorylate -ion of TgGRA7-I regulates its interaction with the PYD domain of ASC, facilitating ASC oligomerization and inflammasome activation | - | Induces proinflammatory responses |
| TgMIF [37] | Triggers NLRP3 inflammasome activation | Hepatocytes | Induces the intense inflammation and pyroptosis, causes fatal liver damage |
| TgMAPKs [38] | Activate NLRP1 and NLRP3 inflammasome, upregulate expression of IL-18, NLRP1/3, ASC, caspase-1, and downregulate expression of IL-10 and IFN-β | Mice | Increase acute virulence |
| TgPrx [39] | Promotes alternatively activated macrophages and reduces caspase-1 activity and IL-1β secretion | Macrophages | Modulates immunopathology and promotes parasite replication and survival |
ASC Apoptosis-associated speck-like protein containing a CARD, IFN interferon, IL interleukin, NACHT nucleotide-binding and oligomerization domain, NF-κB nuclear factor kappa-light-chain-enhancer of activated B cells, NLRP NLR family pyrin-containing protein, PYD pyrin domain, TgGRA T. gondii dense granule protein, TgMAPK T. gondii mitogen-activated protein kinase, TgMIF T. gondii macrophage migration inhibitory factor, TgPrx T. gondii peroxiredoxin, TgROP7 T. gondii Rhoptry protein 7
Toxoplasma gondii profilin induces NLRP3 activation and IL-1β secretion in THP-1 cells [29]. In that study, at 5 h, a decrease in the messenger RNA (mRNA) expression level of the NLRP3 gene was observed, whereas the protein expression of NLRP3 significantly increased after profilin treatment. The release of IL-1β was found to be significantly reduced by treatment with an NLRP3-specific inhibitor [29]. Rhoptry protein 7 of T. gondii (TgROP7) interacts with the NACHT domain of NLRP3, leading to an increase in NF-κB and continuous secretion of IL-1β through the IL-1β/NF-κB/NLRP3 feedback loop in differentiated THP-1 cells. In that study, deficiency of TgROP7 in tachyzoites did not influence parasite replication in host cells but attenuated parasite-induced inflammatory activity [30]. Toxoplasma gondii dense granule protein (TgGRA9) regulates NLRP3 inflammasome activation to treat host sepsis and interacts with NLRP3, inhibiting the formation of the NLRP3 inflammasome by preventing the binding of ASC to NLRP3 in mitochondria. The C-terminus of TgGRA9 is critical for this regulation due to its association with NLRP3, enhancing its anti-inflammatory and bactericidal effects through M2 polarization and exhibiting anti-septic properties [31]. TgGRA15 has been shown to activate the host innate immune response by mediating monocyte activation of the NLRP3 inflammasome and the production of IL-1β [32, 40]. IL-1β in combination with IFN-γ stimulates inducible NOS (iNOS) expression and NO production in hepatocytes, which markedly reduces the production of indole 2,3-dioxygenase 1, a critical IFN-γ-inducible anti-Toxoplasma protein, thereby supporting parasite growth [32]. TgGRA7 and TgGRA15 interact with tumor necrosis factor receptor-associated factors (TRAFs) and activate the NF-κB pathway, strongly inducing NF-κB-mediated proinflammatory responses [33, 35] . Further investigation revealed that TgGRA15-dependent and TgGRA7-dependent NF-κB activation are Myd88-independent and MyD88-dependent, respectively [34, 35]. Protein kinase C alpha (PKCα)-mediated phosphorylation of TgGRA7-I (Ser52) regulates its interaction with the PYD domain of ASC, facilitating ASC oligomerization and inflammasome activation [36]. Toxoplasma gondii macrophage migration inhibitory factor (TgMIF) triggers NLRP3 inflammasome activation in hepatocytes, resulting in pyroptosis. The intense inflammation induced by TgMIF restricts the proliferation of intracellular parasites but causes fatal liver damage. In contrast, TgMIF-deficient parasites significantly attenuate liver injury and prolong mouse survival [37]. The T. gondii mitogen-activated protein kinases TgMAPK1 and TgMAPK2 are associated with NLRP1 and NLRP3 inflammasome activation in mice infected with the virulent T. gondii strain, and parasites lacking TgMAPK1 and TgMAPK2 show reduced acute virulence in mice, characterized by low levels of IL-18, NLRP1/3, ASC and caspase-1 and high levels of IL-10 and interferon beta (IFN-β) transcripts [38]. Treatment with the T. gondii redox enzyme peroxiredoxin (TgPrx) induces alternatively activated murine macrophages and reduces caspase-1 activity and IL-1β secretion in macrophages, and in vitro proliferation of the T. gondii strain RH increases after pretreatment of macrophages with TgPrx [39].
In addition, the ginsenoside Rh2 attenuates neuropathological damage and neuronal apoptosis in the cortical tissue of mice during T. gondii infection. Studies have shown that the ginsenoside Rh2 binds to T. gondii calcium-dependent protein kinase 1 and NLRP3 and decreases the expression of the specific microglial markers ionized calcium-binding adapter molecule 1 and NLRP3 inflammasome-related proteins, leading to the inhibition of the NLRP3 inflammasome and a protective effect against T. gondii infection-induced neuronal injury in microglia [41].
Other inflammasomes
PRU genomic DNA (gDNA) specifically activates the AIM2 inflammasome in peritoneal macrophages, leading to increased susceptibility to T. gondii PRU infection in mice lacking AIM2 [28]. Activation of the AIM2 inflammasome results in the release of caspase-1, mature IL-1β and GSDMD cleavage, contributing to the pyroptosis of placental cells induced by T. gondii [42].
The activation of the immune response and inflammation in toxoplasmosis is associated with NLRP12. In one study, the transcription of the NLRP12, caspase-3, caspase-1, IL-1β, IL-18 and ASC genes was upregulated in nucleated cells within the peritoneal fluid, and a significantly greater level of the NLRP12 protein was observed in the T. gondii-infected BALB/c murine model than in the control group [43].
The role of inflammasome-related genes in the innate immune response to T. gondii infection is crucial. Toxoplasma gondii infection led to time-dependent increases in the mRNA expression of inflammasome sensors such as NLRP1, NLRP3, NLRC4, NLRP6, NLRP8, NLRP13, AIM2, and NAIP, along with elevated mRNA levels of the adaptor proteins ASC, caspase-1, and IL-1β in THP-1 macrophages [44]. Additionally, soluble antigens from T. gondii strain RH tachyzoites time-dependently upregulated NLRP1, NLRP3, NLRC4, and AIM2 gene expression; downregulated IL-1β and IL-18; and increased IL-1β release in THP-1 macrophages [45]. In FHs Int 74 cells infected with T. gondii, the mRNA expression of NOD2, NLRP3, NLRP6, and NAIP1 significantly increased, whereas the expression of NLRP2, NLRP7, and CIITA decreased. Toxoplasma gondii infection activated the NLRP3, NLRP6, and NLRC4 inflammasomes in these cells, resulting in the production of IL-1β, IL-18, and IL-33. Interestingly, NLRP6 inflammasome activation in response to T. gondii was independent of the MAPK pathway in FHs 74 Int cells [10]. Furthermore, T. gondii infection of human placental cells induced the activation of the NLRP1, NLRP3, NLRC4, and AIM2 inflammasomes and increased ASC expression and the levels of mature IL-1β, cleaved caspase-1, and GSDMD, leading to pyroptosis in placental trophoblasts and amniotic cells. The administration of ROS scavengers, a CatB inhibitor, or inflammasome-specific siRNA reversed adverse pregnancy outcomes in a T. gondii-infected pregnant murine model [42]. Moreover, Caspase-8 enhances NF-κB family member IκB kinase phosphorylation, facilitates c-Rel nuclear translocation, and regulates IL-12 and IL-1β expression. Administration of exogenous IL-12 restores monocyte recruitment and effectively controls parasite burden in Ripk3–/–Casp8–/– mice [46]. Further investigation revealed that caspase-8 regulates IL-1β secretion via a mechanism independent of gasdermin in T. gondii-infected human monocytes. Simultaneous pharmacological inhibition of caspase-8 and RIPK1 in primary monocytes reduces IL-1β release without affecting cell viability or parasite infection [47].
Noncanonical inflammasomes
Protective immunity against T. gondii invasion is generated through inflammasome signaling, which suppresses the production of type I IFN (IFN-I). This suppression is crucial for IFN-γ production by natural killer cells and the recruitment of inflammatory monocytes to the infection site [48]. Mechanistically, inflammasome-coupled IL-1β signaling via caspase-1/caspase-11 triggers the expression of the negative regulator SOCS1. SOCS1 binds to INF regulatory factor 3, inhibiting IFN-I production [28]. Additionally, T. gondii-induced activation of the caspase-1/caspase-11-dependent inflammasome and IL-18 is essential for sustaining robust CD4+ TH1 IFN-γ responses in a mouse model of T. gondii infection in the absence of Toll-like receptor 11 (TLR11) [49].
Caspase-11 plays a protective role by increasing inflammation early in infection to mitigate disease severity later in toxoplasmosis. Mice lacking caspase-11 exhibit notably lower cytokine production, reduced pathogenesis and decreased inflammation during the acute phase of T. gondii strain ME49 infection. Conversely, caspase-11 deficiency results in increased neuroinflammation and increased accumulation of tissue cysts in the brain during the chronic phase of infection [50].
In conclusion, the activation of inflammasomes by T. gondii varies depending on the host. In addition to the NLRP1 and the NLRP3 inflammasome, AIM2, NLRC4, NLRP6, NLRP8, NLRP12, NLRP13 and NAIP inflammasomes, as well as noncanonical inflammasomes, have been investigated both in vivo and in vitro during T. gondii infection. Toxoplasma gondii manipulates inflammasome activation and inhibition, influencing immune responses and inflammation. These inflammasomes are crucial for regulating the T. gondii burden, controlling parasite growth and proliferation and managing parasite-induced cell death. Therefore, inflammasomes play a vital role in orchestrating innate immune responses against T. gondii infection.
Plasmodium
Malaria is a lethal infectious disease caused by protozoan parasites of the genus Plasmodium that affects millions of people globally. In 2022, approximately 249 million cases and 608,000 associated deaths were reported in 85 countries [51]. There are four primary human-infecting Plasmodium species, namely Plasmodium ovale, P. malariae, P. vivax, and P. falciparum, of which the most significant are P. falciparum and P. vivax. The infection process begins when hepatocytes are invaded by sporozoites, followed by asexual reproduction within liver cells. Plasmodium must develop and replicate within hepatocytes before progressing to the life-threatening blood stage of the disease. Merozoites, which are released from hepatocytes, periodically infect erythrocytes, leading to malaria-associated pathologies. Typical symptoms include intermittent chills, fever and sweating, potentially resulting in spleen enlargement; anemia; and, in severe cases, damage to vital organs such as the brain, liver and kidneys or even multisystem failure of the circulatory and respiratory systems, ultimately leading to death. Placental malaria is associated with significant inflammation, leading to complications such as abortion, preterm delivery and intrauterine growth restriction. Cerebral malaria is a serious neurological complication caused by P. falciparum infection and is characterized by parasite sequestration, inflammatory cytokine production and vascular leakage. Murine experimental cerebral malaria (ECM) strains have been generated using Plasmodium berghei ANKA (PbA) strain and Plasmodium chabaudi, as malaria parasites infecting humans do not infect mice.
It has been established that innate immune responses in the liver can modulate Plasmodium infection, potentially reducing the incidence and severity of clinical malaria [52–56]. The robust IFN-I response enhances innate immune control, resulting in delayed progression to the blood stage of malaria, reduced parasitemia and increased survival in mice [53, 57]. Thus, inducing type I IFNs to limit Plasmodium infection in the liver decreases the extent and impact of clinical malaria [57].
The rupture of infected erythrocytes triggers an inflammatory response induced by parasitic factors such as Plasmodium PAMPs or DAMPs, which leads to the production of inflammatory cytokines such as TNF alpha (TNF-α), IFN-γ, IL-1β and IL-18. Plasmodium components, including gDNA, RNA, hemozoin (Hz) and glycosylphosphatidylinositol, are captured by immune cells, stimulating the activation of inflammasomes such as AIM2, NLRP3 or NLRC4 and resulting in cytokine release, which manifests as cyclic paroxysm and high fever. Recognition of Plasmodium glycosylphosphatidylinositol by host TLR2 and TLR4, cytosolic RNA by TLR7 and parasite DNA and Hz-DNA complexes by TLR9 has been noted [58–61]. NLRs and AIM2 significantly contribute to inflammasome activation [56]. During Plasmodium yoelii infection, Plasmodium gDNA, RNA and Hz lead to the activation of the AIM2 and the NLRP3 inflammasome in dendritic cells and macrophages [62]. High concentrations of immunocomplexes containing parasite DNA induce the AIM2 and NLRP3 inflammasome assembly, caspase-1 activation and IL-1β secretion in monocytes from infected patients, thereby stimulating systemic inflammation during acute malaria episodes [63]. Systemic inflammation resulting from the parasite often dictates the clinical presentation. Recent studies on the role of host inflammasomes in preventing Plasmodium parasite infection are summarized in the following sections (Fig. 2).
Fig. 2.
Role of inflammasomes in modulating the immune response against Plasmodium infection. The AIM2 and the NLRP3 inflammasome are predominantly activated during the Plasmodium life cycle and the pathogenesis of malaria. In liver cells infected by Plasmodium, Plasmodium DNA is detected by AIM2 receptors in hepatocytes, and ASC facilitates complete caspase-1 processing, enhances GSDMD-mediated pyroptotic cell death and enables the secretion of mature IL-1β and IL-18, thereby improving the overall control of Plasmodium infection in the murine liver [85]. In erythrocytes infected by Plasmodium, when complexed with the malarial pigment hemozoin, Plasmodium DNA induces TLR9 translocation, post-phagocytosis and hemozoin (Hz) and DNA dissociation [64]. DNA and Hz induce AIM2 and NLRP3 inflammasome activation, respectively [64]. Hz activates the NLRP3 inflammasome through Lyn and Syk kinases [68]. Uric acid may enhance the initial host inflammatory response to hemozoin via NLRP3 inflammasome activation [71]. Caspase 8 and the NLRP3 inflammasome regulate host inflammatory responses in cerebral malaria. AIM2 and the NLRP3 inflammasome contribute to the pathology associated with placental malaria [65]. ASC, Apoptosis-associated speck-like protein containing a CARD; GSDMD, N-terminal of gasdermin D; IFN, interferon; IL, interleukin; MDA5, melanoma differentiation-associated protein 5; MAVS, mitochondrial antiviral signaling protein; NLRP, NLR family pyrin-containing protein; TLR, Toll-like receptor
AIM2 inflammasome
The AIM2 inflammasome and its components are involved in the inflammatory responses of Plasmodium-infected hepatocytes, erythrocytes, liver and placenta. In macrophages, plasmodial gDNA on the surface of Hz activates the AIM2 inflammasome and induces mature IL-1β production [64], as already demonstrated in studies of T. gondii infection [28, 42]. In Plasmodium-infected hepatocytes, Plasmodium DNA is detected by cytosolic AIM2 receptors, leading to the activation of inflammasome-mediated caspase-1, GSDMD cleavage and pyroptotic cell death, contributes to the elimination of Plasmodium-infected hepatocytes and the control of malaria infection in the liver [52]. Plasmodium-infected erythrocytes induce AIM2 inflammasome activation and IL-1β production [64]. Furthermore, the activation of the AIM2 inflammasome and caspase-1-dependent signaling negatively regulate host immune responses against lethal P. yoelii YM infection, and parasitemia is reduced in infected AIM2- and caspase-1-deficient mice compared with wild-type mice. Mice lacking AIM2 and caspase-1 are completely resistant to P. yoelii YM infection [62]. The AIM2 inflammasome also affects fetal growth and viability and placental IL-1β production during murine placental malaria and contributes substantially to placental malaria-related pathogenesis [65].
NLRP3 inflammasome
The role of the NLRP3 inflammasome is important in terms of the ability of the host to resist Plasmodium infection at three key parasitic sites: erythrocytes, the brain and the placenta.
Erythrocytes
In Plasmodium-infected erythrocytes, the intraerythrocytic parasite Plasmodium produces Hz, a crystalline detoxification product of hemoglobin, which is released into the circulation during erythrocyte lysis and facilitates further infection of naive red blood cells. The potent proinflammatory effect of Hz through inflammasome activation is implicated in Plasmodium-associated pathologies [66]. Previous studies demonstrated that the malarial pigment Hz transports plasmodial DNA to a subcellular compartment accessible to TLR9, inducing inflammatory signals [64]. The stimulation of TLR9 by natural Hz depends on the presence of Plasmodium DNA on the crystal surface rather on than the Hz itself [67], suggesting a collaborative role of Hz and DNA in inducing systemic inflammation during malaria infection. Hz is inherently immunoreactive, activating the NLRP3 inflammasome and leading to the secretion of IL-1β and IL-18 [57, 64, 66–69]. The inhibition of phagocytosis, potassium efflux and NADPH oxidase activity in THP-1 cells and BMDMs blocks Hz activity [66]. Research has shown that the activation of the NLRP3 inflammasome is induced by Hz via the Src kinase Lyn and the spleen tyrosine kinase Syk in vivo and in vitro [68]. Genetic, chimeric and pharmacological methods have demonstrated that the parasite-derived crystal Hz depletes conventional CD8α+ type 1 dendritic cells and also impairs their function in an NLRP3 inflammasome-dependent manner in vivo, resulting in decreased CD4+ T follicular helper cell differentiation and weakened anti-Plasmodium humoral immunity [70]. Based on these study results, NLRP3 inflammasome-mediated immune recognition and the inflammatory response are considered to be elicited by Hz [66, 71, 72].
Activation signals in addition to Hz, such as uric acid, extracellular ROS production, potassium efflux, and Syk and NADPH oxidase-2 activation, induce NLRP3 inflammasome activation [71, 73, 74]. First, the levels of uric acid, the degradation products of the purine derivatives hypoxanthine and xanthine that accumulate in Plasmodium-infected erythrocytes, are increased in the bloodstream following the rupture of infected red blood cells during severe malaria in humans and mice [75–77]. Uric acid may enhance the initial host inflammatory response to Hz via NLRP3 inflammasome activation, which is characterized by neutrophil recruitment and inflammatory cytokine and chemokine production in vivo. Nevertheless, inflammasome activation by synthetic Hz is mitigated by allopurinol, an inhibitor of uric acid synthesis [71]. In contrast, uric acid does not play a role in NLRP3 activation by synthetic Hz β-hematin in THP-1 cells [66]. Second, heme, a marker of hemolysis or extensive cell damage, activates the NLRP3 inflammasome and promotes IL-1β synthesis in macrophages by inducing Syk and NADPH oxidase-2 activation, mitochondrial ROS production, and potassium efflux, which play critical roles in malaria pathogenesis [73]. Finally, increased plasma xanthine oxidase activity is associated with elevated levels of inflammatory cytokines and the onset of cerebral malaria in malaria patients [74]. Subsequent investigations demonstrated that extracellular ROS generated by xanthine oxidase activate the NLRP3 inflammasome and promote the processing of pro-IL-1β, triggering a potent inflammatory response in primary human monocyte-derived macrophages [74].
Brain
In rodent cerebral malaria, the role of the NLRP3 inflammasome is contentious. One study revealed that mice lacking NLRP3 presented delayed onset of cerebral malaria, whereas those deficient in caspase-1, the adaptor protein ASC, or the IL-1 receptor presented symptoms similar to those of wild-type mice. Thus, in PbA-induced murine cerebral malaria, NLRP3 functions independently of the inflammasome and the IL-1 receptor [78]. Another study revealed that infection of NLRP3-deficient mice with PbA sporozoites suppressed cerebral malaria pathogenesis without affecting parasitemia [66]. Similarly, the absence of NLRP3 or IL-1β resulted in increased survival and lower body temperature in mice during Plasmodium chabaudi adami DS infection [68], demonstrating partial protection to the ECM. Moreover, the activation of the inflammasome-associated components IL-1β and IL-18 through caspase-1 does not influence PbA-induced immunopathology [79]. These studies demonstrate that NLRP3, ASC, caspase-1, IL-18 or the IL-1 receptor have only minor or no effects on the control of parasitemia and the development of the mouse ECM [68, 80], which is contrary to findings in T. gondii [10]. However, mice deficient in NLRP3 and caspase-1 are resistant to P. yoelii YM infection and present lower levels of parasitemia than wild-type mice do, indicating that NLRP3 and caspase-1 inflammasome activation negatively modulates host immune responses to resist lethal P. yoelii YM infection [62].
In one study, the expression of IL-33 at both the gene and protein levels decreased in the brain during fatal ECM [81]. Treatment with IL-33 following PbA-induced murine cerebral malaria reduced NLRP3 inflammasome formation and IL-1β secretion in microglia and intracerebral monocytes during the acute recovery phase. These findings highlight the role of the IL-33-NLRP3 pathway in neuroinflammation and cerebral pathology, which impacts the efficacy of antimalarial drug treatment for cerebral malaria [81].
In brain endothelial cells, NLRP3 mRNA expression is upregulated after exposure to PbA [78]. The virulence of histidine-rich protein II, a diagnostic and prognostic marker for P. falciparum infection, lies in its delivery of numerous hemes to cerebral microvascular endothelial cells, causing iron toxicity, ROS production and activation of the NLRP3 inflammasome, leading to IL-1β production. This sequence disrupts human cerebral microvascular endothelial barrier integrity, increases endothelial permeability and promotes vascular inflammation and edema [79].
Placenta
The NLRP3 inflammasome impairs fetal growth in pregnant mice with placental malaria and may contribute significantly to the development of placental malaria-related pathology. Anakinra, a pharmacological IL-1 receptor antagonist, has shown promise in improving pregnancy outcomes by promoting fetal growth and reducing resorption in experimental placental malaria [65].
Other inflammasomes
Investigations on the impact of single nucleotide polymorphisms on hematological and clinical parameters through multivariate analysis have revealed that the NLRP1 inflammasome receptor is crucial in determining the clinical outcomes of P. vivax malaria, including fever, anemia and thrombocytopenia. IL-1β rs1143634 is notably associated with patient parasitemia, whereas IL-18 rs5744256 appears to play a protective role against anemia development.
The NLRP12 inflammasome is present in monocytes from patients with febrile P. vivax infection, and activation of the NLRP12 inflammasome by Plasmodium is likely a pivotal event in promoting systemic IL-1β production and heightened sensitivity to bacterial superinfection during malaria sepsis [82].
Caspase-11, caspase-4 and caspase-8 are activated in both PbA- and P. chabaudi-infected mice and in patients infected with P. vivax and P. falciparum. Both caspase-1 and caspase-8 are involved in the production and secretion of IL-1β and TNF-α, thereby contributing to increased susceptibility to septic shock and the development of ECM in P. chabaudi-infected mice and P. berghei-induced ECM [83]. In malaria patients, caspase-1, caspase-4, caspase-8 and GSDMD are activated in monocytes [74, 82, 83], which may contribute to the overall increased levels of IL-1β and immunopathogenesis during P. vivax and P. falciparum infections [74, 82, 83]. The IL-18 system is integral to the pathogenesis of P. falciparum, with both IL-18- and IL-18-binding proteins correlating positively with disease severity, parasitemia and endothelial cell activation in the plasma of malaria patients [84].
In the liver, inflammasome-mediated caspase-1 activation further enhances the control of Plasmodium, thus reducing the frequency and severity of clinical malaria. However, caspase-1 processing in both mouse and human hepatocytes is incomplete, preventing the secretion of mature IL-1β or IL-18. This incomplete processing is attributed to the inherently low expression of the inflammasome adaptor molecule ASC in murine hepatocytes. In contrast, transgenic overexpression of ASC in hepatocytes facilitates complete caspase-1 processing, enhances GSDMD-mediated pyroptotic cell death and enables the production of proinflammatory cytokines, such as mature IL-1β and IL-18, which are otherwise absent, thereby improving the overall control of Plasmodium infection in the murine liver [85].
Caspase-12 attenuated parasite clearance in blood-stage malaria and impacted susceptibility to cerebral malaria by limiting immune control of parasite replication and reducing cerebral hyperinflammation in a cerebral malaria model. Caspase-12 competes with a key regulator of NF-κB for binding to IκB kinase-α/β, blocking the formation of the IκB kinase complex and inhibiting downstream NF-κB-dependent transcriptional activation, separate from the caspase-1 inflammasome [86].
Recent evidence suggests that different host inflammasomes are involved in distinct stages of Plasmodium infection. The AIM2 inflammasome regulates Plasmodium control in the liver. In erythrocytes infected by Plasmodium, the NLRP3, NLRP12 and AIM2 inflammasomes participate in immune recognition and inflammatory responses. The NLRP3 inflammasome, caspase-4, caspase-8, caspase-11 and caspase-12 are crucial for regulating host inflammatory responses in cerebral malaria. The AIM2 and the NLRP3 inflammasome contribute to the pathology associated with placental malaria [65]. Inflammasomes play key roles in immune recognition, inflammation, parasite control, the development of Plasmodium-related pathologies and the severity of clinical malaria.
Comparison of the role of inflammasomes in the host response to apicomplexan parasites
The intracellular protozoan parasites Toxoplasma and Plasmodium cause systemic infections in a wide range of cell and tissue types. Multiple inflammasomes, including the AIM2, NLRP1, NLRP3 and NLRP12 inflammasomes and caspase-8, which play critical roles in initiating the inflammatory response to resist Toxoplasma and Plasmodium infections, have been studied. Research on the role of inflammasomes against Toxoplasma and Plasmodium infections has been conducted not only at the cellular level but also in animal models. The NLRP1 inflammasome pathway triggers cellular pyroptosis and directs resistance to toxoplasmosis in Lewis rats, whereas the NLRP1 receptor is crucial in determining the clinical outcomes of P. vivax malaria [87]. Moreover, the NLRP3 inflammasome mediates immune recognition and the inflammatory response and eliminates two pathogens to protect the host, modulating the pathogenesis of Toxoplasma and Plasmodium infections. The role of the NLRP3 inflammasome in rodent cerebral malaria is controversial. The activation signals, including ROS, ATP, P2X7 receptor, potassium efflux and T. gondii molecules, induce NLRP3 inflammasome activation in response to Toxoplasma infection; however, Hz, uric acid and extracellular ROS production; potassium efflux; and Syk and NADPH oxidase-2 activation induce NLRP3 inflammasome activation in the context of Plasmodium infection [64, 71, 73, 74]. Whether the activation of the P2X7 receptor can activate the NLRP3 inflammasome against Plasmodium infection remains unknown. Furthermore, the AIM2 inflammasome, which is activated by the gDNA of two pathogens, contributes to pyroptosis in T. gondii-infected placental cells and is involved in the inflammatory responses of Plasmodium-infected hepatocytes, erythrocytes, the liver and the placenta. Caspase-1 and caspase-8 are involved in the production and secretion of IL-1β during Toxoplasma and Plasmodium infections.
Research on the host inflammasome of Toxoplasma and Plasmodium differs. First, in terms of the types of inflammasomes studied, the NLRP1 and the NLRP3 inflammasome have been studied more in the context of Toxoplasma infection, and the AIM2 and the NLRP3 inflammasome have been studied against Plasmodium infection, with more studies on the NLRP3 inflammasome. Future studies are needed to investigate the role of other inflammasomes. The noncanonical inflammasome caspase-11 plays a protective role in T. gondii infection, promoting an inflammatory response early in infection and reducing disease severity later in infection [50], but the role of noncanonical inflammasomes underlying Plasmodium infection is still unclear. Second, in terms of parasite effector proteins, several molecules in Toxoplasma gondii are involved in NLRP3 inflammasome regulation, GRA9 negatively regulates inflammasome activation, and the rest activate the inflammasome. Plasmodium Hz, DNA and RNA induce inflammasome activation. Further studies are needed to understand the specific role of other parasitic products in NLRP3 inflammasome regulation.
In addition to Plasmodium and Toxoplasma, Cryptosporidium also belongs to the phylum Apicomplexa. There is little research on Cryptosporidium. Epithelial cell-intrinsic NLRP6/caspase-1-mediated secretion of IL-18 is required for the control of Cryptosporidium [88]. Compared with wild-type mice, mice deficient in core components of the inflammasome are more susceptible to Cryptosporidium parvum and C. tyzzeri infections [88, 89]. Research has shown that the NLRP1, NLRP3 and AIM2 inflammasomes are associated with the control of the related apicomplexan parasites Toxoplasma and Plasmodium but are not required for the control of Cryptosporidium. This discrepancy may reflect the pronounced differences in the tissue and host-cell tropisms of apicomplexan parasites, with Cryptosporidium being restricted to enterocytes, whereas Toxoplasma and Plasmodium cause systemic infections in a wide range of cell types [88].
Conclusions
Inflammasomes, recognized as critical signals in innate immunity, control parasite burden, manage parasite-induced cell death and play crucial roles in the onset and progression of toxoplasmosis and malaria. Toxoplasma gondii has evolved multiple strategies to evade host immune defenses; however, the underlying mechanisms of these strategies are still not well understood. Further research is needed on how T. gondii bidirectionally regulates inflammasomes and modulates the magnitude of inflammasome activation and whether other apicomplexan parasites bidirectionally regulate inflammasomes to balance inflammation and promote chronic disease. It is increasingly important to maintain the proinflammatory effects of inflammasomes in a curative rather than pathogenic state during infections. A deeper understanding of how parasitic infections regulate inflammasomes could significantly improve our knowledge of the host immune response to apicomplexan infections. Insights into the regulatory mechanisms of inflammasomes are essential for advancing drug discovery, adjuvant development and vaccine production.
Acknowledgements
Not applicable.
Abbreviations
- ASC
Apoptosis-associated speck-like protein containing a CARD
- BMDMs
Bone marrow-derived cells
- CARD
Caspase-activating and recruitment domain
- DAMPs
Damage-associated molecular patterns
- ECM
Experimental cerebral malaria
- GSDMD
Gasdermin D
- GSDMD-NT
N-terminal of gasdermin D
- Hz
Hemozoin
- IFN
Interferon
- IL
Interleukin
- iNOS
inducible NOS
- LPS
Lipopolysaccharide
- LRR
Leucine-rich repeat
- MAVS
Mitochondrial antiviral signaling protein
- MDA5
Melanoma differentiation-associated protein 5
- mRNA
messenger RNA
- NACHT
Nucleotide-binding and oligomerization domain
- NF-κB
Nuclear factor kappa-light-chain-enhancer of activated B cells
- NLR
Nucleotide binding oligomerization domain-like receptor
- NLRP
NLR family pyrin-containing protein
- NO
nitric oxide
- PAMPs
Pathogen-associated molecular patterns
- PbA
Plasmodium berghei ANKA strain
- PKCα
Protein kinase C alpha
- PRRs
Pattern recognition receptors
- PYD
Pyrin domain
- ROS
Reactive oxygen species
- TgGRA
T. gondii Dense granule protein
- TgMAPK
T. gondii Mitogen-activated protein kinase
- TgMIF
T. gondii Macrophage migration inhibitory factor
- TgPrx
T. gondii Peroxiredoxin
- TgROP7
T. gondii Rhoptry protein 7
- TLR11
Toll-like receptor 11
- TNF-α
TNF alpha
- TRAFs
Tumor necrosis factor receptor-associated factors
Author contributions
Zhi-xin Wang: Conceptualization. Zhi-xin Wang: Writing—original draft preparation. Wan-jun Jiao and Yong Yang: Writing—review and editing. Hai-long Wang and Hong-li Liu: Supervision. All authors have read and approved the published version of the manuscript.
Funding
This study was supported by Fundamental Research Program of Shanxi Province (202203021212365) and Science Research Start-up Fund for Doctor of Shanxi Medical University (XD2112).
Availability of data and materials
No datasets were generated or analysed during the current study.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Contributor Information
Hong-li Liu, Email: lhl0425@sina.com.
Hai-long Wang, Email: longwty@163.com.
References
- 1.Barnett KC, Li SR, Liang KX, Ting JPY. A 360° view of the inflammasome: mechanisms of activation, cell death, and diseases. Cell. 2023;186:2288–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pappas G, Roussos N, Falagas ME. Toxoplasmosis snapshots: global status of Toxoplasma gondii seroprevalence and implications for pregnancy and congenital toxoplasmosis. Int J Parasitol. 2009;39:1385–94. [DOI] [PubMed] [Google Scholar]
- 3.Flegr J, Prandota J, Sovicková M, Israili ZH. Toxoplasmosis—a global threat: correlation of latent toxoplasmosis with specific disease burden in a set of 88 countries. PLoS ONE. 2014;9:e90203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Jeffers V, Tampaki Z, Kim K, Sullivan WJ. A latent ability to persist: differentiation in Toxoplasma gondii. Cell Mol Life Sci. 2018;75:2355–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rostami A, Riahi SM, Gamble HR, Fakhri Y, Shiadeh MN, Danesh M, et al. Global prevalence of latent toxoplasmosis in pregnant women: a systematic review and meta-analysis. Clin Microbiol Infect. 2020;26:673–83. [DOI] [PubMed] [Google Scholar]
- 6.Orchanian SB, Lodoen MB. Monocytes as primary defenders against Toxoplasma gondi infection. Trends Parasitol. 2023;39:837–49. [DOI] [PubMed] [Google Scholar]
- 7.Khan IA, Moretto M. Immune responses to Toxoplasma gondi. Curr Opin Immunol. 2022;77:102226. [DOI] [PubMed] [Google Scholar]
- 8.Lima TS, Gov L, Lodoen MB. Evasion of human neutrophil-mediated host defense during Toxoplasma gondii infection. MBio. 2018;9:e02027-e2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ewald SE, Chavarria-Smith J, Boothroyd JC. NLRP1 is an inflammasome sensor for Toxoplasma gondii. Infect Immun. 2014;82:460–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gorfu G, Cirelli KM, Melo MB, Mayer-Barber K, Crown D, Koller BH, et al. Dual role for inflammasome sensors NLRP1 and NLRP3 in murine resistance to Toxoplasma gondii. MBio. 2014;5:e01117-e1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Cirelli KM, Gorfu G, Hassan MA, Printz M, Crown D, Leppla SH, et al. Inflammasome sensor NLRP1 controls rat macrophage susceptibility to Toxoplasma gondii. PLoS Pathog. 2014;10:e1003927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Matsuno SY, Pandori WJ, Lodoen MB. Capers with caspases: Toxoplasma gondii tales of inflammation and survival. Curr Opin Microbiol. 2023;72:102264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Cavailles P, Flori P, Papapietro O, Bisanz C, Lagrange D, Pilloux L, et al. A highly conserved Toxo1 haplotype directs resistance to Toxoplasmosis and its associated Caspase-1 dependent killing of parasite and host macrophage. PLoS Pathog. 2014;10:e1004005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wang YF, Cirelli KM, Barros PDC, Sangaré LO, Butty V, Hassan MA, et al. Three Toxoplasma gondii dense granule proteins are required for induction of Lewis rat macrophage pyroptosis. MBio. 2019;10:e02388-e2418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wang YF, Hollingsworth LR, Sangare LO, Paredes-Santos TC, Krishnamurthy S, Penn BH, et al. Host E3 ubiquitin ligase ITCH mediates Toxoplasma gondii effector GRA35-triggered NLRP1 inflammasome activation and cell-autonomous immunity. MBio. 2024;15:e0330223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Witola WH, Mui E, Hargrave A, Liu SS, Hypolite M, Montpetit A, et al. NALP1 influences susceptibility to human congenital Toxoplasmosis, proinflammatory cytokine response, and fate of Toxoplasma gondii-infected monocytic cells. Infect Immun. 2011;79:756–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gov L, Schneider CA, Lima TS, Pandori W, Lodoen MB. NLRP3 and potassium efflux drive rapid IL-1β release from primary human monocytes during Toxoplasma gondii infection. J Immunol. 2017;199:2855–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Pandori WJ, Lima TS, Mallya S, Kao TH, Gov L, Lodoen MB. Toxoplasma gondii activates a Syk-CARD9-NF-κB signaling axis and gasdermin D-independent release of IL-1β during infection of primary human monocytes. PLoS Pathog. 2019;15:e1007923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chu JQ, Gao FF, Wu WY, Li CC, Pan ZB, Sun JH, et al. Expression profiles of NOD-like receptors and regulation of NLRP3 inflammasome activation in Toxoplasma gondii-infected human small intestinal epithelial cells. Parasit Vectors. 2021;14:153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lees MP, Fuller SJ, McLeod R, Boulter NR, Miller CM, Zakrzewski AM, et al. P2X7 receptor-mediated killing of an intracellular parasite, Toxoplasma gondii, by human and murine macrophages. J Immunol. 2010;184:7040–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Moreira-Souza ACA, Almeida-da-Silva CLC, Rangel TP, Rocha GD, Bellio M, Zamboni DS, et al. The P2X7 receptor mediates Toxoplasma gondii control in macrophages through canonical NLRP3 inflammasome activation and reactive oxygen species production. Front Immunol. 2017;8:1257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Corrêa G, Lindenberg CDA, de Abreu Moreira-Souza AC, Savio LEB, Takiya CM, Marques-da-Silva C, et al. Inflammatory early events associated to the role of P2X7 receptor in acute murine toxoplasmosis. Immunobiology. 2017;222:676–83. [DOI] [PubMed] [Google Scholar]
- 23.Moreira-Souza ACA, Coutinho-Silva R. The complexity of purinergic signaling during Toxoplasma infection. Curr Top Med Chem. 2021;21:205–12. [DOI] [PubMed] [Google Scholar]
- 24.Quan JH, Huang R, Wang Z, Huang S, Choi IW, Zhou Y, et al. P2X7 receptor mediates NLRP3-dependent IL-1β secretion and parasite proliferation in Toxoplasma gondii-infected human small intestinal epithelial cells. Parasit Vectors. 2018;11:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Moreira-Souza ACA, Nanini HF, Rangel TP, da Silva SRB, Damasceno BP, Ribeiro BE, et al. P2X7 receptor modulation of the gut microbiota and the inflammasome determines the severity of Toxoplasma gondii-induced ileitis. Biomedicines. 2023;11:555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jamieson SE, Peixoto-Rangel AL, Hargrave AC, de Roubaix LA, Mui EJ, Boulter NR, et al. Evidence for associations between the purinergic receptor P2X7(P2RX7) and toxoplasmosis. Genes Immun. 2010;11:374–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Miller CM, Zakrzewski AM, Robinson DP, Fuller SJ, Walker RA, Ikin RJ, et al. Lack of a functioning P2X7 receptor leads to increased susceptibility to toxoplasmic ileitis. PLoS ONE. 2015;10:e0129048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hu ZQ, Wu D, Lu JS, Zhang YF, Yu SM, Xie YC, et al. Inflammasome activation dampens Type I IFN signaling to strengthen anti-Toxoplasma immunity. MBio. 2022;13:e0236122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Pazoki H, Mirjalali H, Niyyati M, Tabaei SJS, Mosaffa N, Shahrokh S, et al. Toxoplasma gondii profilin induces NLRP3 activation and IL-1β production/secretion in THP-1 cells. Microb Pathog. 2023;180:106120. [DOI] [PubMed] [Google Scholar]
- 30.Zhu LJ, Qi WJ, Yang G, Yang YR, Wang YW, Zheng LL, et al. Toxoplasma gondii Rhoptry Protein 7 (ROP7) interacts with NLRP3 and promotes inflammasome hyperactivation in THP-1-derived macrophages. Cells. 2022;11:1630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kim JS, Mun SJ, Cho E, Kim D, Son W, Jeon HI, et al. Toxoplasma gondii GRA9 regulates the activation of NLRP3 inflammasome to exert anti-septic effects in mice. Int J Mol Sci. 2020;21:8437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gov L, Karimzadeh A, Ueno N, Lodoen MB. Human innate immunity to Toxoplasma gondii is mediated by host Caspase-1 and ASC and parasite GRA15. MBio. 2013;4:e00255-e313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bando H, Lee Y, Sakaguchi N, Pradipta A, Ma JS, Tanaka S, et al. Inducible nitric oxide synthase is a key host factor for Toxoplasma GRA15-dependent disruption of the gamma interferon-induced antiparasitic human response. MBio. 2018;9:e01738-e1818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Yang CS, Yuk JM, Lee YH, Jo EK. Toxoplasma gondii GRA7-induced TRAF6 activation contributes to host protective immunity. Infect Immun. 2015;84:339–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sangaré LO, Yang NH, Konstantinou EK, Lu D, Mukhopadhyay D, Young LH, et al. Toxoplasma GRA15 activates the NF-κB pathway through interactions with TNF receptor-associated factors. MBio. 2019;10:e00808-e819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rosowski EE, Lu DN, Julien L, Rodda L, Gaiser RA, Jensen KDC, et al. Strain-specific activation of the NF-κB pathway by GRA15, a novel Toxoplasma gondii dense granule protein. J Exp Med. 2011;208:195–212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Koh HJ, Kim YR, Kim JS, Yun JS, Jang K, Yang CS. Toxoplasma gondii GRA7-targeted ASC and PLD1 promote antibacterial host defense via PKCα. PLoS Pathog. 2017;13:e1006126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhou N, Chen Y, Wang J, An R, Cai HJ, Liu ST, et al. TgMIF promotes hepatocyte pyroptosis and recruitment of proinflammatory macrophages during severe liver injury in acute toxoplasmosis. J Infect Dis. 2023;227:1417–27. [DOI] [PubMed] [Google Scholar]
- 39.Wang SC, Wang ZD, Gu Y, Li ZY, Li ZY, Wei F, et al. Toxoplasma gondii mitogen-activated protein kinases are associated with inflammasome activation in infected mice. Microbes Infect. 2016;18:696–700. [DOI] [PubMed] [Google Scholar]
- 40.Marshall ES, Elshekiha HM, Hakimi MA, Flynn RJ. Toxoplasma gondii peroxiredoxin promotes altered macrophage function, caspase-1-dependent IL-1β secretion enhances parasite replication. Vet Res. 2011;42:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jin GN, Lu JM, Lan HW, Lu YN, Shen XY, Xu X, et al. Protective effect of ginsenoside Rh2 against Toxoplasma gondii infection-induced neuronal injury through binding TgCDPK1 and NLRP3 to inhibit microglial NLRP3 inflammasome signaling pathway. Int Immunopharmacol. 2022;112:109176. [DOI] [PubMed] [Google Scholar]
- 42.Quan JH, Gao FF, Ma TZ, Ye W, Gao X, Deng MZ, et al. Toxoplasma gondii induces pyroptosis in human placental trophoblast and amniotic cells by inducing ROS production and activation of cathepsin B and NLRP1/NLRP3/NLRC4/AIM2 inflammasome. Am J Pathol. 2023;193:2047–65. [DOI] [PubMed] [Google Scholar]
- 43.Rajabi S, Spotin A, Mahami-Oskouei M, Baradaran B, Babaie F, Azadi Y, et al. Toxoplasma gondii activates NLRP12 inflammasome pathway in the BALB/c murine model. Acta Trop. 2022;225:106202. [DOI] [PubMed] [Google Scholar]
- 44.Chu JQ, Shi G, Fan YM, Choi IW, Cha GH, Zhou Y, et al. Production of IL-1β and inflammasome with upregulated expressions of NOD-like receptor related genes in Toxoplasma gondii-infected THP-1 macrophages. Korean J Parasitol. 2016;54:711–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Pazoki H, Rahimi HM, Mirjalali H, Niyyati M, Mosaffa N, Tabaei SJS, et al. Soluble total antigen derived from Toxoplasma gondii tachyzoites increased the expression levels of NLRP1, NLRP3, NLRC4, AIM2, and the release of mature form of IL1β, but downregulated the expression of IL1β and IL18 genes in THP-1cell line. Microb Pathog. 2021;158:105072. [DOI] [PubMed] [Google Scholar]
- 46.DeLaney AA, Berry CT, Christian DA, Hart A, Bjanes E, Wynosky-Dolfi MA, et al. Caspase-8 promotes c-Rel-dependent inflammatory cytokine expression and resistance against Toxoplasma gondii. Proc Natl Acad Sci USA. 2019;116:11926–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Pandori WJ, Matsuno SY, Shin J-H, Kim SC, Kao TH, Mallya S, et al. Role for caspase-8 in the release of IL-1β and active caspase-1 from viable human monocytes during Toxoplasma gondii infection. J Immunol. 2024;212:1161–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Matta SK, Olias P, Huang Z, Wang QL, Park E, Yokoyama WM, et al. Toxoplasma gondii effector TgIST blocks type I interferon signaling to promote infection. Proc Natl Acad Sci USA. 2019;116:17480–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.López-Yglesias AH, Camanzo E, Martin AT, Araujo AM, Yarovinsky F. TLR11-independent inflammasome activation is critical for CD4+T cell-derived IFN-γ production and host resistance to Toxoplasma gondii. PLoS Pathog. 2019;15:e1007872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Coutermarsh-Ott SL, Doran JT, Campbell C, Williams TM, Lindsay DS, Allen IC. Caspase-11 modulates inflammation and attenuates Toxoplasma gondii pathogenesis. Mediators Inflamm. 2016;2016:9848263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.The Lancet Infectious Diseases. A new dawn for malaria prevention. Lancet Infect Dis. 2024;24:107. [DOI] [PubMed]
- 52.Marques-da-Silva C, Poudel B, Baptista RP, Peissig K, Hancox LS, Shiau JC, et al. AIM2 sensors mediate immunity to Plasmodium infection in hepatocytes. Proc Natl Acad Sci USA. 2023;120:e2210181120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Marques-da-Silva C, Peissig K, Walker MP, Shiau J, Bowers C, Kyle DE, et al. Direct type I interferon signaling in hepatocytes controls malaria. Cell Rep. 2022;40:111098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Kurup SP, Anthony SM, Hancox LS, Vijay R, Pewe LL, Moioffer SJ, et al. Monocyte-derived CD11c+ cells acquire Plasmodium from hepatocytes to prime CD8 T cell immunity to liver-stage malaria. Cell Host Microbe. 2019;25:565-577e566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Liehl P, Zuzarte-Luís V, Chan JN, Zillinger T, Baptista F, Carapau D, et al. Host-cell sensors for Plasmodium activate innate immunity against liver-stage infection. Nat Med. 2014;20:47–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Gazzinelli RT, Kalantari P, Fitzgerald KA, Golenbock DT. Innate sensing of malaria parasites. Nat Rev Immunol. 2014;14:744–57. [DOI] [PubMed] [Google Scholar]
- 57.He XA, Xia L, Tumas KC, Wu J, Su XZ. Type I interferons and malaria: a double-edge sword against a complex parasitic disease. Front Cell Infect Microbiol. 2020;10:594621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Barboza R, Lima FA, Reis AS, Murillo OJ, Peixoto EPM, Bandeira CL, et al. TLR4-mediated placental pathology and pregnancy outcome in experimental malaria. Sci Rep. 2017;7:8623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Baccarella A, Fontana MF, Chen EC, Kim CC. Toll-like receptor 7 mediates early innate immune responses to malaria. Infect Immun. 2013;81:4431–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wu J, Tian LJ, Yu X, Pattaradilokrat S, Li J, Wang MJ, et al. Strain-specific innate immune signaling pathways determine malaria parasitemia dynamics and host mortality. Proc Natl Acad Sci USA. 2014;111:E511–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Coban C, Igari Y, Yagi M, Reimer T, Koyama S, Aoshi T, et al. Immunogenicity of whole-parasite vaccines against Plasmodium falciparum involves malarial hemozoin and host TLR9. Cell Host Microbe. 2010;7:50–61. [DOI] [PubMed] [Google Scholar]
- 62.Yu X, Du Y, Cai C, Cai B, Zhu M, Xing C, et al. Inflammasome activation negatively regulates MyD88-IRF7 type I IFN signaling and anti-malaria immunity. Nat Commun. 2018;9:4964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hirako IC, Gallego-Marin C, Ataide MA, Andrade WA, Gravina H, Rocha BC, et al. DNA-containing immunocomplexes promote inflammasome assembly and release of pyrogenic cytokines by CD14+CD16+CD64highCD32low inflammatory monocytes from malaria patients. MBio. 2015;6:e01605-e1615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Kalantari P, DeOliveira RB, Chan J, Corbett Y, Rathinam V, Stutz A, et al. Dual engagement of the NLRP3 and AIM2 inflammasomes by Plasmodium-derived hemozoin and DNA during malaria. Cell Rep. 2014;6:196–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Reis AS, Barboza R, Murillo O, Barateiro A, Peixoto EPM, Lima FA, et al. Inflammasome activation and IL-1 signaling during placental malaria induce poor pregnancy outcomes. Sci Adv. 2020;6:eaax6346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Dostert C, Guarda G, Romero JF, Menu P, Gross O, Tardivel A, et al. Malarial hemozoin is a Nalp3 inflammasome activating danger signal. PLoS ONE. 2009;4:e6510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Parroche P, Lauw FN, Goutagny N, Latz E, Monks BG, Visintin A, et al. Malaria hemozoin is immunologically inert but radically enhances innate responses by presenting malaria DNA to Toll-like receptor 9. Proc Natl Acad Sci USA. 2007;104:1919–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Shio MT, Eisenbarth SC, Savaria M, Vinet AF, Bellemare MJ, Harder KW, et al. Malarial hemozoin activates the NLRP3 inflammasome through Lyn and Syk kinases. PLoS Pathog. 2009;5:e1000559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Corbett Y, Parapini S, D’Alessandro S, Scaccabarozzi D, Rocha BC, Egan TJ, et al. Involvement of Nod2 in the innate immune response elicited by malarial pigment hemozoin. Microbes Infect. 2015;17:184–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Pack AD, Schwartzhoff PV, Zacharias ZR, Fernandez-Ruiz D, Heath WR, Gurung P, et al. Hemozoin-mediated inflammasome activation limits long-lived antimalarial immunity. Cell Rep. 2021;36:109586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Griffith JW, Sun T, McIntosh MT, Bucala R. Pure hemozoin is inflammatory in vivo and activates the NALP3 inflammasome via release of uric acid. J Immunol. 2009;183:5208–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jaramillo M, Bellemare MJ, Martel C, Shio MT, Contreras AP, Godbout M, et al. Synthetic Plasmodium-like hemozoin activates the immune response: a morphology—function study. PLoS ONE. 2009;4:e6957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Dutra FF, Alves LS, Rodrigues D, Fernandez PL, de Oliveira RB, Golenbock DT, et al. Hemolysis-induced lethality involves inflammasome activation by heme. Proc Natl Acad Sci USA. 2014;111:E4110–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ty MC, Zuniga M, Götz A, Kayal S, Sahu PK, Mohanty A, et al. Malaria inflammation by xanthine oxidase-produced reactive oxygen species. Embo Mol Med. 2019;11:e9903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Gallego-Delgado J, Ty M, Orengo JM, van De Hoef D, Rodriguez A. A surprising role for uric acid: the inflammatory malaria response. Curr Rheumatol Rep. 2014;16:401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Lopera-Mesa TM, Mita-Mendoza NK, van de Hoef DL, Doumbia S, Konaté D, Doumbouya M, et al. Plasma uric acid levels correlate with inflammation and disease severity in Malian children with Plasmodium falciparum malaria. PLoS ONE. 2012;7:e46424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Orengo JM, Evans JE, Bettiol E, Leliwa-Sytek A, Day K, Rodriguez A. Plasmodium-induced inflammation by uric acid. PLoS Pathog. 2008;4:e1000013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Reimer T, Shaw MH, Franchi L, Coban C, Ishii KJ, Akira S, et al. Experimental cerebral malaria progresses independently of the Nlrp3 inflammasome. Eur J Immunol. 2010;40:764–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Nguyen ST, Du DN, Wychrij D, Cain MD, Wu QP, Klein RS, et al. Histidine- rich protein II nanoparticle delivery of heme iron load drives endothelial inflammation in cerebral malaria. Proc Natl Acad Sci USA. 2023;120:e2306318120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kordes M, Matuschewski K, Hafalla JCR. Caspase-1 activation of interleukin-1β (IL-1β) and IL-18 is dispensable for induction of experimental cerebral malaria. Infect Immun. 2011;79:3633–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Strangward P, Haley MJ, Albornoz MG, Barrington J, Shaw T, Dookie R, et al. Targeting the IL33-NLRP3 axis improves therapy for experimental cerebral malaria. Proc Natl Acad Sci USA. 2018;115:7404–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ataide MA. Malaria-induced NLRP12/NLRP3-dependent caspase-1 activation mediates inflammation and hypersensitivity to bacterial superinfection. PLoS Pathog. 2014;10:e1003885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Pereira LMN, Assis PA, de Araújo NM, Durso DF, Junqueira C, Ataíde MA, et al. Caspase-8 mediates inflammation and disease in rodent malaria. Nat Commun. 2020;11:4596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Otterdal K, Berg A, Michelsen AE, Yndestad A, Patel S, Gregersen I, et al. IL-18 and IL-18 binding protein are related to disease severity and parasitemia during falciparum malaria. BMC Infect Dis. 2021;21:1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Marques-da-Silva C, Schmidt-Silva C, Baptista RP, Kurup SP. Inherently reduced expression of ASC restricts caspase-1 processing in hepatocytes and promotes Plasmodium infection. J Immunol. 2024;212:596–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Labbé K, Miu J, Yeretssian G, Serghides L, Tam M, Finney CA, et al. Caspase-12 dampens the immune response to malaria independently of the inflammasome by targeting NF-κB signaling. J Immunol. 2010;185:5495–502. [DOI] [PubMed] [Google Scholar]
- 87.Santos MLS, Reis EC, Bricher PN, Sousa TN, Brito CFA, Lacerda MVG, et al. Contribution of inflammasome genetics in Plasmodium vivax malaria. Infect Genet Evol. 2016;40:162–6. [DOI] [PubMed] [Google Scholar]
- 88.Sateriale A, Gullicksrud JA, Engiles JB, McLeod BI, Kugler EM, Henao-Mejia J, et al. The intestinal parasite Cryptosporidium is controlled by an enterocyte intrinsic inflammasome that depends on NLRP6. Proc Natl Acad Sci USA. 2021;118:e2007807118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.McNair NN, Bedi C, Shayakhmetov DM, Arrowood MJ, Mead JR. Inflammasome components caspase-1 and adaptor protein apoptosis-associated speck-like proteins are important in resistance to Cryptosporidium parvum. Microbes Infect. 2018;20:369–75. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No datasets were generated or analysed during the current study.



