Skip to main content

Inflammasomes: potential therapeutic targets in hematopoietic stem cell transplantation

Abstract

The realm of hematopoietic stem cell transplantation (HSCT) has witnessed remarkable advancements in elevating the cure and survival rates for patients with both malignant and non-malignant hematologic diseases. Nevertheless, a considerable number of patients continue to face challenges, including transplant-related complications, infection, graft failure, and mortality. Inflammasomes, the multi-protein complexes of the innate immune system, respond to various danger signals by releasing inflammatory cytokines and even mediating cell death. While moderate activation of inflammasomes is essential for immune defense and homeostasis maintenance, excessive activation precipitates inflammatory damage. The intricate interplay between HSCT and inflammasomes arises from their pivotal roles in immune responses and inflammation. This review examines the molecular architecture and composition of various types of inflammasomes, highlighting their activation and effector mechanisms within the context of the HSCT process and its associated complications. Additionally, we summarize the therapeutic implications of targeting inflammasomes and related factors in HSCT.

Introduction

Hematopoietic stem cell transplantation (HSCT) entails the intravenous infusion of hematopoietic stem and progenitor cells (HSPCs) after conditioning regimens like high-dose chemotherapy and/or radiotherapy. The primary objective is to eradicate residual tumour cells via graft-versus-leukemia (GVL) effects, while simultaneously replacing damaged or dysfunctional HSPCs to rejuvenate both the hematopoietic and immune systems [1]. Since the pioneering efforts of Gatti, Good [2], and Thomas [3] in the late 1960s, who initially employed HSCT to address human immunodeficiency disorders and aplastic anemia, this technology has been widely adopted for treating benign and malignant hematopoietic diseases. As a curative approach, HSCT has significantly improved clinical outcomes. From 1957 to 2019, over 1.5 million HSCT procedures were performed worldwide, with an annual growth rate exceeding 10% [4]. Despite recent significant advancements in HSCT, such as expanded indications, improved management of complications, and better integration with targeted therapies and immunotherapies [5]. HSCT still poses substantial risks for graft failure, relapse, and severe transplant-related complications, which are closely linked to immune tolerance and reconstruction [6, 7]. Therefore, elucidating the mechanisms underlying these adverse outcomes and identifying novel therapeutic targets are crucial.

The innate immune system acts as the body’s initial defense mechanism against invasion and endogenous threats, playing a critical role in eliminating infections and damaged cells [8]. The central mechanism of this immune response is triggered by cytoplasmic protein complexes called inflammasomes [9]. Inflammasomes possess a remarkable capacity to swiftly discern pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). Their activation subsequently triggers innate immune responses by facilitating the release of pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18), initiating an inflammatory cascade [10]. Proper inflammasome activation can swiftly eliminate damaged cells and strengthen immune defense. However, prolonged activation causes severe tissue damage in the body and may induce pyroptosis, a highly inflammatory form of cell death characterized by cellular lysis and destruction [11]. More than ten types of inflammasomes have been identified currently, each intricately intertwined in the pathogenesis of inflammatory and metabolic diseases, including cancer, diabetes, and infections [12, 13]. The intense reactivity of inflammasomes to various danger signals broadens their association with other diseases. Emerging evidence supports that inflammasomes are widely expressed in the hematopoietic system and have participated in several critical stages of HSCT. NLRP1, NLRP2, and NLRP3 have demonstrated significant prognostic implications for overall survival, relapse, and non-relapse mortality rates in HSCT [14]. Thus, meticulous modulation of inflammasome activation and tempering their excessive response are beneficial for post-HSCT outcomes.

In this paper, we present an overview of the individuality and commonality among different inflammasomes, focusing on their composition, activation, and effector functions. We systematically address the key roles of inflammasomes in coordinating mobilization, hematopoietic reconstitution, and associated complications, with particular emphasis on acute graft-versus-host disease (aGVHD). Finally, we assess the potential benefits of targeting inflammasomes in the treatment of HSCT.

Inflammasomes

Classification and composition of inflammasomes

The concept of inflammasomes, first described by Martinon et al. (2002), refers to multiprotein complexes and immune regulators that respond to pathogenic or physiological stimuli [10]. To date, more than ten types of inflammasomes have been identified, distinguished primarily by their components, activation mechanisms, and biological functions. Inflammasomes are categorized into three types according to their constituent proteins: the NLR family, the ALR family, and the Pyrin [15]. This section introduces several common inflammasomes.

The NLR family

The NLR family is the most well-characterized, consisting primarily of NLRP1, NLRP3, NLRP6, and NLRC4 [16]. These inflammasomes feature three functionally distinct domains. The C-terminal leucine-rich repeat (LRR) domain acts as the “sensor”, recognizing and binding ligands due to its unique three-dimensional structure [17]. These ligands include exogenous PAMPs and endogenous DAMPs, differing in their molecular origins. The innate immune system rapidly recognizes and responds to signals from invading pathogens or damaged self-cells by distinguishing between self and non-self components [8]. The nucleotide-binding domain (NBD), also referred to as NACHT, is shared by other NLR members and facilitates LRR oligomerization by binding adenosine triphosphate (ATP). This interaction relieves the autoinhibition of LRR and promotes immune signal transduction [18]. Due to the sensitivity of the NBD to mutations, functional changes are commonly observed [19]. It is worth mentioning that NLRP1 exerts its function independently of the ATPase activity of the NBD. It features a unique FIIND domain non-covalently linked to the N-terminus to self-cleavage, separating the N-terminal and C-terminal domains of NLRP1 and leading to its activation [20]. Another significant protein domain is the variable N-terminal domain containing the pyrin domain (PYD), caspase recruitment domain (CARD) as well as baculovirus inhibitor of apoptosis repeat (BIR) domain. These domains interact with the adaptor protein ASC through PYD-PYD or CARD-CARD linkages, ultimately resulting in the activation of pro-caspase-1 [21]. NLRs are categorized into four subfamilies based on their divergent N-terminal domains: CIITA with an acidic transactivation domain, BIR domain, NLRPs with an N-terminal Pyrin domain and NLRCs with an N-terminal CARD domain [16].

The ALR family

The ALR family includes AIM2 and IFI16, both of which structurally contain the C-terminal oligonucleotide DNA-binding HIN-200 domain and an N-terminal domain composed solely of PYD [22]. These domains facilitate the nonspecific recognition of DNA through electrostatic interactions, where positively charged consecutive oligonucleotide/oligosaccharide-binding residues in the HIN domain interacts with the negatively charged sugar-phosphate backbone of double-stranded DNA (dsDNA). This electrostatic interaction enables the HIN domain to recognize aberrant endogenous or exogenous DNA structures [23]. Subsequently, the signal is relayed to the N-terminal PYD, which exhibits a pronounced tendency for self-oligomerization. Binding of dsDNA induces conformational changes in the HIN domain, exposing the PYD and initiating signal transduction [24]. The exposed PYD interacts with ASC, promoting oligomerization and recruiting caspase-1 via CARD-CARD interactions [25]. Unlike AIM2, the IFI16 protein can localize to both the nucleus and the cytoplasm. IFI16 contains two HIN-200 domains, which are separated by a linker region rich in serine, threonine, and proline residues [26]. In addition to its role in classical inflammasome activation, the PYD of IFI16 indirectly activates the stimulator of interferon genes (STING) pathway through oligomerization, leading to the induction of type I interferon production [26].

The pyrin

Pyrin, named after the Greek word for “fever”, contains an N-terminal PYD, a B-box domain, a coiled-coil domain, and a C-terminal B30.2/SPRY domain [27]. The B30.2/SPRY domain in Pyrin detects alterations in the cytoskeleton and transmits danger signals, typically triggered by pathogen infections (Fig. 1B) [28]. The PYD interacts with ASC through signal transduction, participating in the activation of the classical inflammasome. The remaining domains contribute to signal transmission and maintaining structural stability [27].

Activation of inflammasomes

The differentiation between priming and activating signals in inflammasomes minimizes redundancy, thereby effectively regulating the immune protective functions of these complexes and mitigating tissue damage under various conditions.

NLRP3, the most extensively studied inflammasome, is activated by a wide range of irritants and pathogens. Its activation requires a dual-signal mechanism to ensure stringent regulation. The first signal, priming, is elicited by both PAMPs and DAMPs. PAMPs encompass conserved structures found on pathogens, such as lipopolysaccharide (LPS) derived from gram-negative bacteria, components of cell walls and viral nucleic acids [29]. DAMPs are endogenous molecules released in response to cellular stress or injury within the host. These include high mobility group box 1 (HMGB1), S100 calcium-binding protein, heat shock proteins, ATP, and uric acid [30]. Both PAMPs and DAMPs are recognized by cell surface receptors such as Toll-like receptors (TLRs) and NOD-like receptors (NLRs), including NOD1 and NOD2 as well as cytokine receptors like the IL-1 receptor and tumor necrosis factor-alpha receptor (TNF-α). The activation of these receptors triggers signaling molecules myeloid differentiation primary response 88 (MyD88), which subsequently lead to the transcriptional activation of NF-κB and the upregulation of NLRP3 [31, 32]. The priming step is regulated by both the transcriptional control of inflammasome components, which modulates the quantity of inflammasome subunits, and post-translational modifications, such as ubiquitination, phosphorylation, and succinylation [33]. Subsequently, the activation signal directly induces the assembly of NLRP3 oligomers, ASC, and pro-caspase-1 into the inflammasome protein complex. This signal is associated with various cellular disturbances, including alterations in ion channels, such as potassium ion (K+) efflux and calcium ion (Ca2+) influx, as well as the generation of reactive oxygen species (ROS) resulting from oxidative stress, mitochondrial dysfunction, lysosomal rupture, and metabolic changes [34]. Collectively, these closely related, multi-layered signals regulate and determine NLRP3 activation.

Anthrax lethal toxin (LeTx) enters the cytoplasm by oligomerizing protective antigen on the host cell membrane. The lethal toxin protease subunit subsequently cleaves the N-terminus of murine NLRP1, thereby exposing its CARD domain and activating the NLRP1 inflammasome [35]. Similar effects are observed with viral proteases that cleave human NLRP1 [36]. Additionally, viral proteases [37], phosphorylation modifications due to viral infection [38], and intracellular DNA damage [39] can alter the conformation of NLRP1. NLRP6 is primarily expressed in the intestines, lungs, and liver, and it plays a crucial role in the host’s defense against microbial infections. Similarly, full activation of the NLRP6 inflammasome requires both priming and activation signals. Priming signals encompass microbial signals (interferon signaling) [40] and metabolic pathways (peroxisome proliferator-activated receptor gamma, PPAR-γ) [41], which promote the transcription and expression of NLRP6 [42]. Once NLRP6 has accumulated sufficiently, activation signals, including microbe-associated molecular patterns (MAMPs), become effective. These MAMPs consist of microbial components and metabolic products, such as lipoteichoic acid (LTA) from gram-positive bacteria [43], LPS and ATP from gram-negative bacteria [44], as well as certain microbial metabolites like taurine, histamine, and spermidine [45]. Additionally, NLRC4 primarily responds to bacterial flagellin and the type III or type IV secretion systems (T3SS or T4SS) of gram-negative bacteria [46]. AIM2 and IFI16 primarily recognize intracellular dsDNA through electrostatic interactions mediated by HIN-200 domain [47]. AIM2 is predominantly activated by pathogen-derived dsDNA, thereby protecting the host from microbial invasion, a phenomenon frequently observed during viral infections [48]. In the non-classical activation pathway, low concentrations of bacterial DNA stimulate type I interferon production via the cyclic GMP-AMP synthase (cGAS) pathway, which serves as a sensor for cytosolic dsDNA. This signaling cascade results in the transcription of interferon-inducible genes, including GTPases, which promote bacteriolysis and facilitate the release of bacterial DNA into the cytoplasm. The released bacterial DNA is subsequently detected and recognized by the AIM2 inflammasome, leading to its activation [49, 50]. IFI16 senses both dsDNA and ssDNA, recruiting the endoplasmic reticulum protein STING. This interaction subsequently activates interferon-gamma (INF-γ) and pro-inflammatory cytokines through the TBK1-IRF3 and NF-κB signaling pathways, thereby exerting significant antiviral effects (Fig. 1A) [51].

Effects of inflammasomes

Despite variations in composition and signaling pathways, inflammasomes exhibit structural similarities that align with their extensive functional overlap. Inflammasomes primarily recruit and activate pro-caspase-1, which then promotes the maturation and secretion of pro-inflammatory cytokines IL-1β and IL-18, and induces cell death through pyroptosis, apoptosis, and PANoptosis [52]. Specifically, inflammasomes begin to assemble upon receiving priming and activation signals, with ASC functioning as an “adapter” or “bridge”. ASC contains PYD and CARD domains, which facilitate interactions through homotypic PYD–PYD and CARD–CARD associations with pro-caspase-1 [53]. Notably, ASC amplifies the inflammasome signaling [54] and is essential for the assembly of most characterized inflammasomes including NLRP3, AIM2, IFI16, and Pyrin. Conversely, NLRP1 and NLRC4, endowed with inherent CARD domains, can interact autonomously with pro-caspase-1 independent of ASC [55].

The assembly of this complex facilitates the proteolytic self-cleavage of pro-caspase-1 at Gly315, resulting in the generation of the active caspase-1 subunits, referred to as p20 and p10 subunits [56]. Once activated, caspase-1 cleaves the downstream effector molecules pro-IL-1β and pro-IL-18, thereby releasing their active forms (IL-1β and IL-18) extracellularly, which initiate a robust inflammatory response to combat the invasion of exogenous pathogens [57]. Additionally, caspase proteins activated by inflammasomes trigger pyroptosis, a distinct programmed cell death pathway characterized by rapid plasma membrane rupture due to the cleavage of pore-forming effector proteins from the gasdermin family [58, 59]. Pyroptosis is further classified into the caspase-1-mediated canonical signaling pathway and the caspase-4/5/11-mediated non-canonical signaling pathway. In the classical pathway, the gasdermin protein, typically GSDMD, is cleaved by caspase-1 at a flexible linker, which releases the self-inhibitory N-terminal pore-forming domain and the C-terminal repressor domain [60]. The N-terminus of specific proteins can bind to particular lipids, such as phosphatidylinositol found in eukaryotic cell membranes and cardiolipin present in prokaryotic or mitochondrial membranes. In certain instances, these interactions may result in oligomerization and the formation of pore structures within the membrane [61].

The influx and efflux of K+, sodium ions (Na+), and water through stable pores initiate various GSDMD-dependent processes, including cell swelling, rapid plasma membrane rupture, and the modulation of pro-inflammatory components such as the cytokines IL-1β and IL-18, intracellular components (antigens or DAMPs), and intracellular hydrolases [62]. Pyroptosis leads to rapid cell failure and creates a potent inflammatory environment, which aids in the defense against infection or malignant transformation. The non-canonical signaling pathway operates independently of inflammasome signaling cascades to activate caspases. Instead, the CARD domains of caspase-4/5/11 directly detect lipopolysaccharides (LPS) within host cells, facilitating the cleavage of GSDMD [63]. Although these caspases can induce pyroptosis, they cannot directly cleave pro-IL-1β or pro-IL-18, necessitating the synergistic activation of canonical inflammasomes to amplify the inflammatory response [64]. Furthermore, there exists a complex interplay between inflammasomes and apoptosis, which includes the cell fate switch among apoptosis, necroptosis, and pyroptosis, as well as the activation or inhibition of inflammasomes by apoptotic pathways [65, 66]. This interplay may also encompass PANoptosis, a composite form of cell death that integrates features from multiple death pathways [67]. A comprehensive discussion of this area exceeds the scope of this study. These multilayered regulatory issues will not be explored in detail here (Fig. 1C).

Given the diversity of inflammasomes and their complex activation and regulatory mechanisms, it is of great importance to delineate the boundaries between moderate activation and excessive activation. The boundaries are dynamically continuous and closely related to the outcomes following activation [68]. Firstly, different activation products influence the nature of inflammasome activation. Generally, PAMPs are regarded as lower-risk and more defensible threats, while cytoplasmic DAMPs can trigger cell death and pose a higher risk [69]. For example, inflammasomes can swiftly detect exogenous bacterial or viral infections and subsequently secrete IL-1β and IL-18 to alert neighboring cells, thereby limiting the spread of infection. Concurrently, inflammasomes can induce pyroptosis, releasing pathogens and destroying their replication niche, which enables neutrophils to rapidly engulf and eliminate the pathogens [70]. This transient and rapidly diminishing immune defense is often advantageous to the host, as it not only clears harmful stimuli but also initiates the healing process to repair tissue damage. However, the relationship between host defense and uncontrolled inflammation is intricate and multifaceted [71]. Excessive activation of inflammasomes can lead to acute inflammatory damage or be exploited by pathogens for immune evasion, but such occurrences are relatively rare [72]. In contrast, the safety window for inflammasome activation in response to DAMPs is narrower, and even a slight imbalance can result in sustained pathological damage. This phenomenon has been found to be closely associated with modern lifestyle factors, including smoking, obesity, and psychological stress, all of which are directly or indirectly linked to the production of DAMPs. These factors may be one of the causes behind the development of various chronic diseases, such as type 2 diabetes and atherosclerosis [69]. Additionally, the activation of inflammasomes in various diseases demonstrates a dual-edged sword effect [73]. On one hand, inflammasome activation facilitates the clearance of pathogens and mitigates infection-induced damage. On the other hand, in autoimmune or metabolic diseases, chronic inflammasome activation may exacerbate disease progression, resulting in fibrosis and sclerosis [70]. In tumors characterized by high heterogeneity, these effects are particularly pronounced. It enhances anti-tumor immunity and slows tumor progression by activating pyroptosis and regulating the activity of natural killer (NK) cells and T cells. However, IL-1β and IL-18 can promote tumor signaling, thereby inducing tumor proliferation, invasion, and angiogenesis [74, 75]. The dual nature of this property is significantly influenced by the interplay of genetic background, tumor microenvironment, tissue type, and downstream molecules. Currently, the boundary between moderate activation and excessive activation remains ambiguous. Research has indicated that epigenetic regulation such as DNA methylation, histone modifications and post-translational modifications, may offer multi-layered insights into the intricate regulation of inflammasomes [76]. Nevertheless, critical issues such as the levels, spatial ranges, intensities, and synergies of inflammatory factors require further investigation. This is particularly important for conducting layered research utilizing cell fate maps, in vivo imaging technology, and organoid models to comprehensively analyze the regulatory network of the inflammasomes.

Fig. 1
figure 1

Inflammasomes are multiprotein complexes composed of several domains with distinct functions. They are activated upon detecting various danger signals, such as pathogen toxins, nucleic acids, secretion systems, changes in cellular structures, metabolic alterations, ionic disturbances, and organelle damage. The PYD domain of inflammasomes interacts with the adaptor protein ASC, facilitating the assembly of the inflammasome complex. Subsequently, the CARD domain of ASC recruits pro-caspase-1 to form the complex. In the canonical signaling pathway, inflammasome activation results in the self-cleavage and activation of caspase-1. Activated caspase-1 then cleaves pro-IL-1β and pro-IL-18 into their mature, biologically active forms. Additionally, it cleaves GSDMD, generating its active N-terminal domain, which oligomerizes to form stable pores in the plasma membrane. This pore formation induces cell swelling and rupture, leading to inflammatory cell death and the release of intracellular substances such as IL-1β, IL-18, HMGB1, and LDH. In the non-canonical signaling pathway, LPS directly binds to the CARD domain of caspase-4/5/11, resulting in the cleavage of GSDMD. However, GSDMD requires NLRP3 activation to cleave pro-IL-1β and pro-IL-18

Role of inflammasomes in hematopoietic stem cell transplantation

Participation in mobilization, homing, and engraftment

Mobilization and homing are critical yet contrasting stages essential for successful HPSCs migration, particularly in the contexts of bone marrow adhesion and chemotaxis [77]. Previous studies have underscored the paramount importance of inflammasomes, especially NLRP3, in maintaining the delicate equilibrium between these processes.

Mobilization can be divided into three distinct phases: initiation, amplification, and effector. The initiation phase is driven by various pro-inflammatory mediators that accumulate through positive feedback mechanisms. Granulocyte-colony stimulating factor (G-CSF) and AMD3100, which are complementary mobilizing agents, are commonly employed to stimulate the production of DAMPs that induce sterile inflammation. These DAMPs activate innate immune cells, primarily neutrophils and monocytes in the microenvironment. Upon tissue damage, these cells release additional DAMPs, such as ATP and ROS [78]. Prior to this cascade, innate immune cells have already recapitulated the initial signal of the NLRP3 inflammasome through the gut-derived LPS from gram-negative bacteria by binding to TLR4 [79, 80]. Subsequently, ATP emerges as the most critical second signal, released extracellularly through the pannexin-1 membrane channel, which serves as an essential transmembrane pathway connecting the intracellular and extracellular environments [81]. Studies showed that inhibition of the pannexin-1 channel by 10Panx impaired the mobilization of HSPCs and negatively affected the engraftment of white blood cells and platelets [82]. In the extracellular space, extracellular ATP (eATP) serves as an activating ligand for several ionotropic P2X and metabotropic P2Y purinergic receptors [83, 84]. Specifically, eATP interacts with purinergic receptors P2X7, P2X4, and P2X1, which are highly expressed in HSPCs, facilitating Ca2+ influx as an activation signal for NLRP3 [85]. Notably, eATP is also processed by cell surface ecto-nucleotidases CD39 and CD73 into its metabolites: ADP and AMP (produced by CD39), and adenosine (produced by CD73) [86]. Conversely, adenosine exerts an inhibitory effect during mobilization through heme oxygenase 1 (HO-1) [87] and inducible nitric oxide synthase (iNOS) [88], suppressing NLRP3 activation and reducing adhesion capacity, ultimately inhibiting the migration of HSPCs [89]. The release of mature and biologically active IL-1β and IL-18 is contingent upon the inflammasome-mediated activation of caspase-1, which facilitates communication among innate immune cells via autocrine and paracrine pathways. This process further stimulates the release of additional DAMPs, such as HMGB1 and S100A9 [90]. It is plausible that NLRP1, AIM2, and NLRP12 inflammasomes may also exert similar effects in response to eATP and other DAMPs; however, the specific mechanisms underlying these interactions remain to be elucidated [91]. Moreover, supplementation with IL-1β or IL-18 demonstrated limited mobilization in NLRP3-deficient and caspase-1-deficient models, a deficiency that could be corrected by the injection of a DAMP mixture (eATP + HMGB1 + S100A9) [92]. These factors appeared to establish a positive feedback loop that enhances sustained ATP release and NLRP3 activation, ultimately leading to optimal mobilization outcomes (Fig. 2A).

During the amplification phase, these DAMPs activate the complement cascade (ComC) through three main pathways: C1q (classical pathway), mannose-binding lectin (MBL) and Factor B (via the alternative pathway) [93]. Notably, DAMPs primarily exert their effects by activating the MBL pathway [94]. The DAMPs-MBL complex subsequently triggers mannose-binding lectin-associated serine proteases (MASPs), which cleave and activate complement 3 (C3) and prothrombin, thereby inducing the activation of ComC and the coagulation cascade (CoaC) [93, 95]. Furthermore, the study found that Factor B deficiency inhibited NLRP3 and the activation of the alternative pathway, leading to poor cell homing and engraftment [96]. Furthermore, the study found that Factor B deficiency inhibited NLRP3 and the activation of the alternative pathway, leading to poor cell homing and engraftment [92]. These pathways converge in a shared terminal response, resulting in the formation of C5a, C3a, and C5b-9 (membrane attack complex, MAC) as potent inflammatory effector molecules, which connect complement, coagulation, and fibrinolysis proteolytic cascades [97, 98] (Fig. 2B).

In the effector phase, the migration of HSPCs is dependent on ligand-receptor interactions between stromal cell-derived factor 1 (SDF-1) and its receptor, C-X-C chemokine receptor type 4 (CXCR4) [99], as well as very late antigen-4 (VLA-4) and its receptor vascular cell adhesion molecule 1 (VCAM-1) [100, 101]. Upon binding to their respective receptors, these agents activate HSPCs and enhance their proliferation. NLRP3 was activated by C5a, desArgC5a, and MAC, maintaining a sterile inflammatory state in the bone marrow microenvironment. These fragments and anaphylatoxins facilitate granulocyte degranulation, leading to the release of hydrolases or lipases, thereby compromising the bone marrow niche associated with the SDF-1/CXCR4 and VLA-4-VCAM-1 axis. Simultaneously, innate immune cells migrate between the bone marrow endothelium and the periphery in response to chemotactic signals [102, 103]. Additionally, a sphingosine-1 phosphate (S1P) gradient transition between bone marrow and peripheral blood induces HSPCs to cross the vascular endothelium and enter peripheral blood. S1P is released by red blood cells upon MAC induction in peripheral blood to facilitate optimal HSPC mobilization [104]. C3a maintains the retention of HSPCs rather than mobilization by enhancing their responsiveness to SDF-1 in the bone marrow to prevent an uncontrolled release of HSPCs [105].

Lipid rafts, which are membrane microdomains enriched in glycosphingolipids and protein receptors, enhance the efficiency of signal transduction and prolong the surface residency of receptors such as CXCR4 and VLA-4. This augmentation increases the sensitivity and responsiveness of HSPCs to microenvironmental signals [106]. The Nox2-ROS-NLRP3 inflammasome axis played a crucial role in regulating lipid raft assembly and the expression of lipid-synthesizing enzymes [97, 107]. eATP and inflammasomes facilitated the incorporation of the homing receptor CXCR4 into lipid rafts, thereby strengthening the response to the SDF-1 gradient and promoting the migration and engraftment of HSPCs (Fig. 2C) [108].

Fig. 2
figure 2

Role of inflammasomes in the three stages of HSCT Mobilization. A In the initiation phase, mobilizing agents stimulate the transmembrane protein pannexin-1 in innate immune cells, leading to the release of eATP. eATP subsequently binds to P2X ion channels (P2X1, P2X4, P2X7), causing an influx of K+ and an efflux of Ca2+, which triggers the activation of inflammasomes and caspase-1. This process also results in the release of various DAMPs, such as IL-1β, IL-18, HMGB1, and S100A9 through GSDMD, thereby creating a positive feedback loop that amplifies inflammation. B During the amplification phase, DAMPs modulate the MBL/MASPs pathway and the alternative complement pathway, triggering the activation of the ComC and CoaC. This results in the production of C3a, C5a, desArgC5a, and the C5b-C9 complex. At this stage, eATP is converted into adenosine by CD39 and CD73, which inhibits mobilization by upregulating HO-1 and iNOS. Pathways promoting mobilization are indicated by red arrows, while the inhibitory pathways associated with adenosine are marked by blue arrows. C In the effector phase, C5a initiates NLRP3 activation in HSPCs, further escalating the inflammatory response. Additionally, these complement molecules enhance granulocyte degranulation, leading to the secretion of hydrolases that weaken the CXCR4-SDF-1 and VLA-4-VCAM-1 axis. S1P is released by the MAC to chemotax HSPCs into peripheral blood vessels. Moreover, lipid rafts are stimulated by increased membrane assembly via the Nox2/ROS/NLRP3 pathway. This process facilitates the integration of CXCR4 and VLA-4 on the plasma membrane, enhancing HSPCs’ sensitivity to environmental signals, thereby promoting their mobilization, homing, and engraftment

Disruption of hematopoiesis and post-transplant hematopoietic reconstruction

Hematopoietic reconstruction is a meticulously orchestrated process wherein transplanted HSPCs engraft, proliferate, and differentiate into various blood cells within the recipients. Successful HSC engraftment is characterized by the clinical recovery of myeloid (neutrophils), erythroid cells and megakaryocyte (platelet) lineages. Recovery standards include meeting hematologic criteria for engraftment and confirming that the hematopoietic cells originate from the donor, a process that typically spans several weeks [109]. In this context, we elucidate the impact of inflammasomes on hematopoiesis and post-transplant hematopoietic reconstruction using the model of “soil (bone marrow microenvironment), environment (external interventions), and seed (HSPCs)” [110].

The bone marrow microenvironment comprises various stromal cells, extracellular matrix components, reticular cells, other connective tissues, and multiple hematopoietic regulatory factors [111]. Bone marrow mesenchymal stem cells (BMSCs) function as self-renewing precursor cells of the microenvironment, and are capable of differentiating into diverse stromal cells. They play dual roles in signal transduction and transcriptional regulation supporting and repairing the microenvironment [112]. Research in NLRP1 knockout mouse transplant models revealed that the multi-lineage differentiation capacity of BMSCs was impaired, with reduced lipid droplets and osteogenic mineralization areas observed microscopically. Meanwhile, the reconstruction of HSPCs was found to be enhanced, particularly within erythroid lineages [113]. Adipogenesis adversely impacts the hematopoietic niche by compressing the hematopoietic space and suppressing hematopoietic signals [114]. While osteogenesis promotes the growth of hematopoietic cells, it may also lead to excessive differentiation, thereby inhibiting long-term hematopoiesis [112]. Another study revealed that Caspase-3 and NLRP3 share similarities in regulating the balance of BMSCs in hematopoiesis. Deficiencies in either Caspase-3 or NLRP3 result in myeloid hematopoiesis characterized by the expansion of myeloid cells (CD11b+Gr-1+ cells), ultimately distorting the normal composition of hematopoietic cells. This phenomenon was associated with a diminished secretion of hematopoietic maintenance factors, such as stem cell factor (SCF) and chemokine C–X–C motif chemokine 12 (CXCL12), which propelled HSPCs towards myeloid differentiation from their quiescent state. Interestingly, Caspase-3-deficient models indirectly influenced the hematopoietic microenvironment by reducing NLRP3 expression with dysregulated release of SCF and CXCL12, rather than directly interfering inflammatory pathways [115]. Smoking has been identified as a detrimental factor in hematopoiesis. Cigarette extracts induced ROS production in MSCs, activating the NLRP3 pathway. Concurrently, gene expression profiles were modified, resulting in the upregulation of inflammation and oxidative stress-related genes and downregulation of genes supportive of hematopoiesis. Collectively, these factors compromised hematopoietic support, as evidenced by the restricted expansion of CD34+CD90+ HSPCs and their diminished multipotency. This detrimental effect was alleviated by the application of ROS and NLRP3 inhibitors, which restored hematopoietic cell engraftment [116]. In the bone marrow microenvironment, a class of substances known as neuropeptides, including substance P (SP) and neurokinin A (NK-A), participate in the “neuro-immune-hematopoietic” communication network [117]. These neuropeptides exhibit antagonistic effects, influencing the expression of HMGB1 and thereby regulating hematopoietic balance. Specifically, SP promoted hematopoiesis by inhibiting HMGB1, while NK-A exerted an opposing effect. HMGB1 had dual roles in hematopoiesis, as it regulated the proliferation of hematopoietic progenitor cells while also preventing excessive proliferation and depletion of HSCs, thus maintaining a balance in long-term hematopoietic and immune functions. Furthermore, the effects of HMGB1 appeared to be lineage-dependent; its blockade enhances T cell subsets and NK cells while simultaneously reducing B cell differentiation [118].

HSPCs, as the foundational elements of hematopoiesis, possess the ability for self-renewal and exhibit multi-directional differentiation potential [119]. Inflammasomes function as regulators of HSPC quantity and differentiation pathways. In NLRP1 knockout mice, a reduction in the expression of pro-inflammatory cytokines IL-18 and IL-1β was noted, alongside decreased inflammatory infiltration in the bone marrow and an increase in hematopoietic cells, particularly red blood cells and platelets. Furthermore, the secretion of adhesion molecules such as VCAM-1, ICAM-1, and E-selectin was elevated, thereby further promoting HSPC proliferation, differentiation, and engraftment [113]. The knockout or inhibition of T-cell protein tyrosine phosphatase (TC-PTP), a widely expressed molecule across various hematopoietic lineages, enhances HSC numbers in both the bone marrow and peripheral blood by modulating the expression of immune factors through dephosphorylation [120]. Notably, TC-PTP deficiency activated the IL-18/IL-18 bp signaling axis, which triggered the production of IL-12 and IFN-γ, thereby promoting rapid expansion of HSPCs [121]. However, another study indicated that elevated IL-18 levels were associated with an increased risk of non-relapse mortality following transplantation [122]. In terms of differentiation potential, inflammasomes initiated the effector protein caspase-1, which cleaved and degraded GATA binding protein 1 (GATA-1), thereby impeding the development of erythroid progenitors and ultimately reducing erythropoiesis. This suggests that artificially regulating inflammasome activity may present a therapeutic avenue for addressing rare hematopoietic imbalance disorders [123]. Similarly, Caspase-3/7/8 exhibited comparable phenomena in this context [124]. NLRP1 also regulated erythroid-myeloid lineage decisions in HSPCs through the phosphorylation and activation of the ZAKα/P38 axis, with inhibition of NLRP1 promoting erythroid differentiation [125]. Moreover, inflammasomes regulated cell fate through metabolic pathways that are essential for the formation of embryonic HSPCs [126]. Glucose metabolism in macrophages produced ROS and hypoxia-inducible factor 1-alpha (HIF1α), which initiated inflammasome activation and sustained IL-1β production. Stimulation of the inflammasome increased the number of multilineage hematopoietic colony-forming units and T-cell progenitors in zebrafish embryos. Additionally, NLRP3 suppressed erythroid differentiation during the embryonic stage while promoting a bias towards lymphoid and myeloid lineages, effectively counteracting embryonic hypoxia [127].

External factors, inclusive of conditioning, immune suppression, infections, and inflammation, primarily exert indirect effects by modulating of HSPCs and the bone marrow microenvironment. Conditioning regimes can deteriorate the bone marrow microenvironment, concomitantly increasing inflammatory infiltration. Following total body irradiation (TBI) preconditioning, there was an upregulation of various inflammasome components, containing NLRP1 and NLRP6, with IL-1β and IL-18 functioning as key mediators of inflammatory damage. Caspase-1 inhibitors demonstrated efficacy in reducing inflammation and promoting the reconstruction of megakaryocytes and other hematopoietic cells [128]. AIM2 detected radiation-induced dsDNA breaks, subsequently recruited ASC domains, and induced caspase-1-dependent pyroptosis in response to ionizing radiation-induced damage [129]. Similarly, ionizing radiation activated the NLRP3/caspase-1 axis in macrophages, resulting in the release of lactate dehydrogenase (LDH) and inflammatory mediators from membrane rupture [130]. NLRP12 played a beneficial role independent of inflammasomes by suppressing TNF and NF-κB signaling during radiation exposure and heat-induced emergency hematopoiesis. This suppression diminished apoptosis in hematopoietic progenitor cells, bolstered the reconstruction of myeloid hematopoietic cells and peripheral immune function, mitigating lung dysfunction resulting from bacterial infections. Throughout this process, the key components of the inflammasome (caspase-1, ASC, IL-1R) remained unchanged [131]. GSDME, an alternative executor of pyroptosis, mediated HSPC swelling, blebbing, and membrane permeabilization in the presence of cisplatin. GSDME deficiency also heightened sensitivity to apoptosis. These findings underscored the role of GSDME in influencing the hematopoietic function of lymphoid and myeloid cells by regulating the balance between apoptosis and pyroptosis under adverse conditions [132].

The dual role of inflammasomes in GVHD

GVHD is the main complication and a leading contributor of non-relapse and transplant-related mortality in allogeneic HSCT, with an incidence of approximately 50% [133]. High-dose corticosteroids, which serve as the first-line therapy for aGVHD, are associated with limited efficacy and considerable toxicity, potentially affecting post-transplant relapse rates and immune reconstitution. Notably, around half of patients with aGVHD demonstrate a lack of response to steroid therapy, leading to poor outcomes and underscoring the urgent need for novel biomarkers and therapeutic strategies [134]. The pathophysiology of aGVHD can be delineated into three stages: the initiation phase, T-cell activation, and effector stages [135].

The initial stage involves tissue damage induced by conditioning regimes, which acts as a catalyst for widespread tissue damage by releasing DAMPs, PAMPs, numerous inflammatory and chemotactic factors [136]. These signals upregulate the expression of major histocompatibility complex (MHC) antigens, adhesion receptors, and other molecules on host antigen-presenting cells (APCs), thereby amplifying the responses of alloreactive donor T cells. Additionally, these danger signals can enter the circulatory system, resulting in systemic GVHD [137]. Subsequently, T cells expressing tissue-specific adhesion molecules and chemotactic receptors migrate towards and infiltrate targeted organs. The second stage is initiated after activation of donor T cells by host APCs [138]. In response to various cytokines and environmental factors, CD4+ T cells can differentiate into specialized effector subtypes, including T helper types 1/2/17, and regulatory T cells [139]. Th1 cells enhance the function of cytotoxic T cells by secreting IFN-γ and TNF-α, and they are recognized as the primary subtype promoting GVHD. Th17 cells, which secrete IL-17, IL-21, and IL-22, primarily target extracellular pathogens and contribute to the tissue damage associated with GVHD. In contrast, Th2 cells regulate humoral immune responses through the secretion of IL-4, IL-5, and IL-13, typically playing a protective role that mitigates GVHD. Overall, Th2 and regulatory T cells (Tregs) are associated with the attenuation of GVHD, while Th1 and Th17 cells are linked to the acceleration of its development and progression [140]. Altering the Th cell landscape helps the immune system prevent inappropriate activation. Cytotoxic effects and inflammatory cytokines drive the progression to the third stage of GVHD. Effector cells, including CD4+ T cells, CD8+ T cells, and NK cells, can exhibit cytotoxic effects through Fas-FasL, perforin/granzyme, and TNF pathways [141]. During this stage, various types of Th cells also release inflammatory cytokines, further contributing to the disease process.

Initiation

PAMPs, DAMPs and cytokines activate APCs, releasing additional alarm signals that sustain and amplify GVHD [137]. Koehn et al. [142] demonstrated that ATP release induced by conditioning regimens activated NLRP3 through binding to the P2X7 receptor. The subsequent release of the inflammatory cytokine IL-1β led to dysfunction of myeloid-derived suppressor cells (MDSCs), compromising their anti-inflammatory efficacy in the microenvironment. The study also revealed that, compared to localized ATP control or selective inhibition of P2X7 receptor and NLRP3 activity (via gene knockout or co-infusion of regulatory T cells to suppress MDSC inflammasome activity), the use of NLRP3 small molecule inhibitors combined with MDSC infusion in mice did not yield the anticipated results. This outcome may be attributed to the systemic inhibition of NLRP3 inflammasomes affecting other immune functions. Another study indicated that NLRP3 activation established a highly inflammatory environment, whereas NLRP3 knockout reduced levels of IL-1β and TNF-α, decreased ATP release, and lowered P2X7 receptor expression [143]. Given the significance of P2X7 in HSCT, 16 SNPs in the P2X7 gene were analyzed in DNA samples from 453 allo-HSCT donors and recipients, revealing correlations between these polymorphisms and clinical outcomes. The study identified that two loss-of-function P2X7 SNPs (Ile568Asn and Glu496Ala) were associated with higher aGVHD incidence and lower aGVHD risk, respectively, thus contributing to the development of personalized clinical prevention strategies [144]. After conditioning with TBI, busulfan/cyclophosphamide (BU/CY), or fludarabine/cyclophosphamide (FLU/CY) regimens, significantly elevated uric acid levels released by damaged cells were observed in the peritoneal cavity, which induced NLRP3 activation and IL-1β production. IL-1β further amplified inflammatory damage and promoted Th17 cell differentiation to exacerbate GVHD. In the early stages, uricase-mediated degradation of uric acid mitigated NLRP3 activation and the inflammatory response [145]. S100A8, S100A7, and S100A9 have been pinpointed as inflammasome-activating molecules, specifically expressed in the saliva of GVHD patients. These proteins might act as endogenous TLR4 ligands, enhancing inflammasome activation while increasing neutrophil binding affinity and leukocyte extravasation [146]. At this stage, inflammasomes complete the establishment of an inflammatory and damaging environment, facilitating the activation and migration of APCs to lymph nodes, where they further activate donor T cells (Fig. 3A).

Activation

The highly inflammatory environment established during the initiation phase lays foundation for subsequent T-cell activation. Host APCs present processed antigen fragments on their surfaces via HLA molecules. When these fragments interact with the T-cell receptor (TCR) domains of donor naive T cells, they rapidly stimulate T-cell proliferation, polarization, and the release of additional cytokines [147]. Inflammasomes play a role in modulating the behavior of APCs. For instance, dendritic cells (DCs) showed potently impaired migration towards inflammatory environments when lacking microRNA-155 in the presence of LPS and ATP chemotaxis. This impairment reduced inflammasome activation, subsequently inhibiting inflammatory events such as ERK, Caspase-1, and IL-1β activation, thereby mitigating the pathological manifestations of GVHD [148]. At this stage, inflammasomes were primarily involved in the differentiation of activated T cells and the subsequent propagation of inflammation. Caspase-11 directly bound to LPS released by microbes, triggering a non-canonical pyroptosis pathway, leading to cell rupture and the release of pro-inflammatory cytokine IL-1α. This cytokine recruited neutrophils and amplified T-cell proliferation. The polarization of Th1 and Th17 cells, along with the production of IFN-γ were also associated with Caspase-11 activation, collectively accelerating the inflammatory response [149]. Elevated levels of HMGB1 have been detected in the serum of patients with aGVHD post-transplant [150]. HMGB1 reduced DNA methylation of the signal transducer and activator of transcription 3 (STAT3) in CD4+ T cells to increase STAT3 mRNA levels in aGVHD patients. This process promoted the induction of promoting Th17 lymphocyte induction and inhibited Treg cell differentiation [151]. The choline metabolite trimethylamine N-oxide (TMAO) activated the NLRP3 inflammasome through initiation signals (NF-κB) and activation signals (mitochondrial ROS), promoting M1 macrophage polarization. These macrophages exhibited higher levels of pro-inflammatory cytokines (e.g., IL-1β, IL-6, TNF-α) and enhanced antigen-presenting capacity, further driving Th1 and Th17 cell polarization, culminating in intensified inflammatory responses and tissue damage. The NLRP3 inhibitor CY-09 significantly attenuated this phenomenon [152]. Furthermore, CD4+ and CD8+ T cells exhibiting elevated levels of active caspase-1 demonstrated pronounced inflammatory transcriptional characteristics and a metabolic phenotype resembling that of myeloid immune cells. This phenotype was marked by the upregulation of pro-inflammatory cytokines and a metabolic shift from oxidative phosphorylation to aerobic glycolysis. Such metabolic reprogramming typically resulted in enhanced metabolic activation, similar to what was in tumor cells, to meet increased energy demands [153]. During this process, these cells also contributed to mitochondrial dysfunction and the release of ROS and mitochondrial DNA, which further activated inflammasomes and intensified the vicious cycle of inflammation (Fig. 3B) [154].

Effector

In the third stage, the ongoing recruitment of both innate and adaptive immune cells, coupled with cytokine stimulation, prompts T cells to exert their effects. The ASC protein, a component of the inflammasomes, serves as a crucial link between the “sensor” and caspase-1 [53]. Cheong et al. [155] have demonstrated that ASC harbored substantial therapeutic potential in GVHD, independent of inflammasome presence or the microbiome. ASC deficiency reduced granzyme B secretion and degranulation in CD8 + T cells. This lowered cytotoxicity and apoptosis, ultimately suppressing GVHD. ASC deficiency impaired GVL function but enough to provide antiviral protection. Inflammasomes amplified the function of effector cells, which in turn, activate them through unique mechanisms. Studies found that CTLs secreted perforin to create transient pores in the APC membrane, leading to granzyme release and ion perturbations, including K+ efflux and Ca2+ influx. These disturbances facilitated the maturation of NLRP3 and IL-1β, which activated CD4+ T cells and CTLs. The study elucidated a positive feedback loop between CTLs and APCs in GVHD and detailed the mechanisms by which adaptive immunity bolstered innate immunity [156]. Moreover, IL-18, a downstream effector of inflammasomes, enhanced the function of effector cells. Notably, in 60% of aGVHD patients, IL-18 and sIL-2R levels culminated on day 10 post-transplant, preceding the clinical manifestation of GVHD (averaging day 15 post-HSCT). This suggested that serum IL-18 levels were valuable indicators of aGVHD, potentially related to its enhancement of Th1 immune responses and CTL induction [157]. The IL-18Rα-neutralizing monoclonal antibody reduced inflammation by interfering with the IL-18/IL-18R interaction, affecting Th1, Th2, and Th17 subpopulations in the peripheral blood of aGVHD animal models. It also inhibited mitogen-activated protein kinase (MAPK) p38 activity and Fas/FasL expression to reduce apoptosis and ameliorate the GVHD response [158].

Mucosa-associated invariant T cells (MAIT cells), innate T cells with specialized antimicrobial functions, are abundant in the gut [159]. During intestinal GVHD, MAIT cells recognized gut bacterial metabolites (e.g., riboflavin) presented by MHC class I-related protein 1 via their TCR and rapidly migrated to the damaged area. Upon activation, MAIT cells proliferated in response to gut flora through the MR1/TCR-dependent pathway involving CD3/CD28, as well as through cytokines via the non-TCR-dependent pathway, including IL-12 and IL-18. Under TCR signaling, MAIT cells secreted more IL-17 to inhibit inflammation and limit CD4+ T cell proliferation. This phenotype shift under IL-12/IL-18 stimulation leads to the secretion of higher amounts of granzyme B, TNF-α, and IFN-γ. This change promotes the expression of additional cytotoxic factors in mature subsets, thereby balancing the GVHD promotion with GVL effects (Fig. 3C) [160].

Double-edged sword

The gastrointestinal tract is one of the most prevalent and lethal sites afflicted by aGVHD [161]. NLRP6 plays a double-edged sword role in maintaining gut homeostasis. On the positive side, it effectively sustains the balance and diversity of the intestinal flora, thereby preserving intestinal integrity through the regulation of inflammatory pathways. By negatively regulating ZAP-70 signaling in donor T cells, NLRP6 alleviated the severity of GVHD. ZAP-70, an essential cytoplasmic tyrosine kinase, promotes T cell proliferation and differentiation through phosphorylation after binding with the TCR [162]. In mouse models, the absence of NLRP6 increased CD4+ T cell proliferation, inhibited apoptosis and promoted pro-inflammatory Th1 cell differentiation by enhancing ZAP-70 phosphorylation. In contrast to the wild-type cells, NLRP6 deficiency did not influence CD8+ T cell cytotoxicity or Treg suppression, indicating that NLRP6 reduces the severity of GVHD while preserving effective GVL responses [163]. NLRP6 played a protective role in the early stages following HSCT. Its absence promoted caspase-3-mediated apoptosis, leading to intestinal epithelial cell death, reduced expression of the tight junction protein occludin, loss of goblet cells, and decreased levels of antimicrobial peptides Reg3γ and Pla2g2a. NLRP6 deficiency also exacerbated inflammation, as evidenced by elevated inflammatory markers, including CD11b and myeloperoxidase, as well as an increased infiltration of inflammatory cells, such as macrophages, DCs, and neutrophils, within the intestinal [164]. Intriguingly, a recent study offered a new perspective, suggesting that NLRP6 deficiency also triggered compensatory inflammasome activity by activating NLRP3 and other inflammatory pathways. The loss of NLRP6 negatively regulated both the NF-κB and MAPK p38 signaling pathways. In NLRP6-deficient mice, significant increases in the expression levels of NLRP3, pro-caspase-1, p20, IL-1β, and IL-18, accompanied by more severe liver damage, inflammatory cell infiltration, and liver fibrosis. It was hypothesized that while NLRP6 activation resulted in the release of moderate levels of IL-1β to eliminate pathogens, NLRP6 deficiency led to excessive NLRP3 activation, thereby causing liver damage due to an intensified inflammatory response [165]. Paradoxically, some studies have reported opposing findings, indicating that NLRP6 deficiency in non-hematopoietic tissues mitigated the severity of aGVHD, particularly in the gastrointestinal tract, leading to reduced tissue damage and improved survival rates. This effect might be attributed to the expression of specific pro-inflammatory factors, activation of immune cells, and the regulation of the gut barrier (e.g., goblet cells, mucin-2, and tight junction proteins) which operated independently of microbiome regulation [166, 167].

Additionally, NLRP3, previously regarded as a positive regulator of GVHD, has been found to have a protective role in non-hematopoietic tissues, particularly in intestinal epithelial cells. G protein-coupled receptors (GPR109A, GPR43, and GPR41) serve as microbial sensors that are essential for maintaining gut homeostasis and safeguarding gut barrier function [168]. Short-chain fatty acids (SCFAs), the bacterial metabolites, acted as ligands for these receptors and moderately activate NLRP3 in non-hematopoietic tissues through extracellular signal-regulated kinase (ERK) phosphorylation. This activation increased the secretion of the gut-protective factor IL-18, thereby supporting epithelial stability [169]. Moreover, SCFAs promoted the production of Tregs, contributing to regulatory and immunosuppressive effects. Nonetheless, this protective mechanism was disrupted when gut symbiotic bacteria were inhibited by the administration of broad-spectrum antibiotics [170].

Fig. 3
figure 3

Correlation between inflammasomes and HSCT. A Initially, conditioning regimens lead to extensive tissue damage throughout the body, triggering substantial PAMPs and DAMPs. These molecules activate inflammasomes in APCs and other cell types, amplifying inflammatory signals by enhancing the function of APCs. Additionally, they suppress anti-inflammatory cells and release further DAMPs. Inflammasomes also impact the structural integrity of intestinal cells and regulate the secretion of antimicrobial peptides. B In the second phase, donor T cells are activated after interacting with APCs. Inflammasomes affect the migration and antigen-presenting abilities of APCs. They also regulate T cell responses by promoting the maturation of CD8+ T cells and driving CD4+ T cells towards a pro-inflammatory phenotype, accelerating the progression of GVHD. C During the third phase, effector cells execute their functions through various molecular mechanisms. CD8+ T cells induce cell damage via Fas-Fas ligand (FasL), perforin/granzyme, and TNF pathways. The cytotoxic effect of CD8+ T cells is further enhanced by inflammasomes. Moreover, inflammasomes influence the proliferation and differentiation of CD4+ T cells and regulate the secretion of pro-inflammatory cytokines, contributing to the development of GVHD

Exacerbation of other post-transplant complications

Infection ranks as one of the leading causes of death following HSCT, primarily due to the severe immunodeficiency [171]. Immunosuppression or immune deficiency renders patients susceptible to pathogenic infections [172]. During the pre-engraftment phase, which spans from conditioning until 30 days post-HSCT, patients experience severe immunosuppression characterized by neutropenia, mucosal damage, and catheter-related risks. The most common pathogens during this period are bacteria, particularly those causing sepsis from gram-positive bacteria, as well as fungal infections (e.g., Candida and Aspergillus) and viral infections (e.g., herpes simplex virus). The early post-engraftment stage, occurring between 30 and 100 days post-transplant, is primarily marked by cellular immune deficiency. Common infections during this phase include those caused by bacterial pathogens (notably gram-negative bacteria), viral infections (such as Cytomegalovirus), and fungal infections (e.g., Aspergillus). In the late post-engraftment stage, which extends beyond 100 days, the immune system gradually recovers, but the main infection risks include invasive fungal infections (e.g., Cryptococcus), viral reactivation (e.g., cytomegalovirus, varicella-zoster virus), and Epstein-Barr virus-associated lymphoproliferative disorders [173, 174]. Despite the limited research on the interplay between infectious factors and inflammasomes during HSCT, it is plausible that inflammasomes, as sentinels of innate immunity, play a crucial role at different stages of transplantation [12]. Pathogens, including bacteria, fungi, viruses, and parasites, are recognized by host cells through their cell wall components, DNA, and metabolic products, which then activate inflammasomes. The extent of activation varies based on the environment and infectious agents involved. For instance, human dsDNA viruses, such as cytomegalovirus, activated the AIM2 inflammasome, associated with higher cell death rates due to the release of IL-18 and IL-1β. AIM2 also interferes with the transcription of early and late viral genes (UL54 and UL83), inhibiting viral replication and promoting cell death to curb viral spread [175]. Studies have demonstrated that virulent strains of HSV-1 increased the expression of NLRP3, NLRP12, and IFI16 inflammasomes. More virulent HSV-1 strains elicited activation of inflammasomes, resulting in a significant accumulation of inflammatory monocytes and neutrophils, which exacerbated disease severity. Notably, this phenomenon was exclusively observed in HSV-positive cells [176]. In HSV-1-infected microglial cells, the NLRP3 inflammasome triggered Gasdermin D-dependent pyroptosis, where the formation of membrane pores promoted the release of cellular contents and exacerbated tissue damage [177]. Among fungal infections, Candida albicans is recognized as one of the most commonly invasive species in HSCT. The transition of yeast cells to the hyphal form was a critical trigger for NLRP3 inflammasome activation. The hyphal form secreted Candidalysin, which damaged the cell membrane and directly activated the NLRP3 inflammasome, leading to the maturation of IL-1β and potentially limiting the invasion of Candida albicans in the host, possibly by disrupting K+ balance [178]. In a model of acute lung injury mediated by Pseudomonas aeruginosa in HSCT, elevated levels of prostaglandin E2 (PGE2) led to increased IL-1β, which depends on the activation of cAMP response element-binding protein (CREB) transcription factor through cAMP mediated by EP2 and EP4 signaling [179]. The production of IL-1β was associated with the activation of caspase-1 in the canonical pathway and caspase-8 in the alternative pathway. Additionally, PGE2 inhibited the activation of autophagosomes LC3 and p62. The combined effect of IL-1β production and limited autophagic protection collaborated to inflict lung damage after infection, which was mitigated by the PGE2 inhibitor indomethacin [180]. Some pathogens have evolved mechanisms to evade host detection [181]. The pathogenic yeast Candida albicans evaded macrophages by inducing NLRP3 inflammasome-dependent pyroptosis, which caused candida lysin-dependent membrane perforation or hyphal membrane piercing. The GSDMD inhibitor Necrosulfonamide effectively mitigated the escape of Candida albicans and alleviated infection symptoms without interfering with caspase-1 cleavage activity [182]. In summary, moderate inflammasome activation can restrict pathogen infection, whereas excessive activation may initiate an inflammatory cascade that clears pathogens while simultaneously causing tissue damage.

In addition to GVHD and infections, inflammasomes also contribute to other post-transplant complications. Pre-transplant conditioning regimens can lead to severe hepatic complications. A cohort study involving pediatric patients who underwent allogeneic HSCT found a significant association between the donor’s IL-1β−511 TT genotype and the cumulative incidence of grade III-IV hepatic veno-occlusive disease (VOD) within three months post-transplant, with rates of 25 ± 13.1% for the TT genotype compared to 2.9 ± 2.9% and 3.6 ± 3.6% for the CT and CC genotypes, respectively (P = 0.024) [183]. Increased expression of NLRP3 in the liver was observed in hepatitis following Bu/Cy treatment, resulting in heightened inflammatory infiltration. Pharmacological NLRP3 inhibitors, such as BAN11-7082, showed potential in reversing this damage and restoring liver function [184]. Disseminated intravascular coagulation (DIC) following transplantation is a relatively rare complication. In a study of 12 patients administered with recombinant human soluble thrombomodulin (rhTM), rhTM was found to reduce levels of fibrinogen degradation products, C-reactive protein, and HMGB1. The mechanism might involve rhTM binding to HMGB1 DNA-binding proteins via its N-terminal lectin-like domain, which inhibited the release of HMGB1, thereby preventing the inflammatory cascade and protecting vascular endothelial cells [185]. Engraftment syndrome (ES) is a clinical syndrome that arises during neutrophil recovery [186]. A comparison of plasma samples from 56 pediatric patients across three groups, including ES, aGVHD, and a combination of both, revealed that those with isolated ES exhibited significantly elevated levels of pro-inflammatory cytokines (IL-1β, IL-6, and IL-12). Notably, the levels of IL-1β and IL-12 were significantly increased, with IL-1β showing a more pronounced rise compared to those with aGVHD [187]. However, discordant findings from another study [188] highlighted the necessity for cautious interpretation of these results.

Therapeutic targeting of inflammasomes in the transplantation process

Given the significance of inflammasomes in HSCT and their intricate molecular mechanisms for detecting and countering danger signals through precise and coordinated assembly, various strategies targeting inflammasomes have shown considerable therapeutic potential in animal models [189]. These strategies are especially promising for preventing GVHD while preserving GVL effects (Table 1).

Table 1 Mechanisms of targeting inflammasomes in HSCT

Numerous studies have focused on targeting the NLRP3 inflammasome through its dual-signal pathway. In a liver injury model induced by BU/CY conditioning, the administration of the NLRP3 inhibitor BAN11-7082 after HSCT selectively inhibited IκB-α phosphorylation and blocked NF-κB release, consequently reducing levels of IL-1β, IL-18, and neutrophil infiltration in liver tissue and ultimately protecting liver function [184]. Berberine, a traditional Chinese medicine utilized for gastrointestinal diseases, has been shown to inhibit the TLR4 signaling pathway and suppress the transmission of the priming signal of NLRP3, effectively reducing the severity of GVHD in the lungs, liver, and colon. Notably, it reversed the impairment of tight junction proteins in the mouse colon and augmented the abundance of beneficial bacteria [190]. The P2X7 receptor, a ligand-gated ion channel that responds to ATP stimulation, facilitates K+ efflux, thereby activating the second signal of NLRP3 [191]. Brilliant Blue G, a P2X7 receptor inhibitor, inhibited NLRP3 activation and reduced macrophage and neutrophil infiltration, with inflammatory mediators such as CXCL8 and CCL2 in peripheral blood decreased, thereby improving liver function compared to untreated mice [192]. Antagomir-155, an inhibitor of microRNA-155, regulates multiple aspects of both innate and adaptive immune responses [193]. Inhibition of miR-155 was reported to decrease the expression of purinergic receptors in DCs, which reduced sensitivity to LPS and ATP stimulation, weakened ERK activation, and decreased inflammatory tendencies of DCs, further inhibiting NLRP3 activation [148].

In addition to the broad-spectrum inhibition of NLRP3, targeting its downstream activator protein, caspase-1, provides greater specificity. The caspase-1 specific inhibitor Ac-YVAD-cmk effectively suppressed the infiltration of macrophages and neutrophils, as well as the production of chemokines. Furthermore, it facilitated hematopoietic reconstruction, leading to an increase in megakaryocytes and platelets observed in post-transplant models [128]. Another study demonstrated that Ac-YVAD-cmk mitigated pathological damage and inflammation in the liver, lungs, and colon by modulating the Th1/Th17/Treg balance and reducing their characteristic pro-inflammatory states. Specifically, it inhibited Th1/Th17 differentiation and promoted Tregs, thereby alleviating damage caused by T cells activation in GVHD [194].

Regulating the expression of downstream inflammatory effectors presents significant therapeutic potential. Antibodies that target IL-18Rα inhibited the interaction between IL-18 and its receptor, thereby affecting the levels of Th1, Th2, and Th17 subpopulations. Specifically, this intervention reduces Th1 and Th17 cell expression while enhancing anti-inflammatory effects. Additionally, these antibodies decreased the expression levels of apoptosis-related molecules such as Fas and FasL, as well as the phosphorylation of MAPK p38, which collectively reduced apoptosis and contributed to the preservation of organ function [158]. Although IL-18 might exert more adverse effects in GVHD treatment, its combination with other cytokines to pre-activate NK cells in vitro prior to in vivo administration demonstrated remarkable therapeutic potential. This cytokine cocktail therapy encompassed two strategies: the IL-12/15/18 and the IL-12/18 methods, both of which pre-activated and matured NK cells with persistent memory retention. NK cells did not directly kill activated T cells or inhibit APCs, instead, they produced IFN-γ and other anti-inflammatory factors, effectively preserving GVL effects. Compared to the IL-12/15/18 method, the IL-12/18 approach offered greater advantages in cell activation, proliferation, and retention. Additionally, IL-12/18 demonstrated sustained beneficial effects in mild aGVHD, indicating enhanced safety [195, 196]. The study revealed that IL-1β, originated from the activation of NLRP3 in intestinal cells, acted on the IL-1 receptors of DCs, CD4+ T cells, and CD8+ T cells. Anakinra, an IL-1 receptor antagonist (IL-1Ra) inhibitor, influenced the polarization of Th17 cells, ultimately improving the survival rate of mice in the early stages of aGVHD [145]. However, a randomized clinical trial displayed that blockade of IL-1 using IL-1Ra during conditioning was not sufficient to reduce GVHD or to improve survival [197].

Other classic upstream inflammasome mediators, including HMGB1 and MyD88, also exhibit excellent inhibitory potential. NecroX-7 inhibited HMGB1 secretion and TLR4 expression in a dose-dependent manner by suppressing mitochondrial ROS and protein kinase C (PKC) pathways, which further modulated the Th1/Treg ratio, reducing inflammatory responses [198]. MyD88, a critical adapter in innate immune signal transduction, transmits signals from TLR and IL-1R family receptors by recruiting interleukin-1 receptor-associated kinase 4 (IRAK4) to activate the NF-κB pathway and inflammasomes. The IRAK4 inhibitor PF-06650833 suppressed MyD88’s function, inhibiting the differentiation of pro-inflammatory cells (Th1, Th17, and Tc1) in GVHD, while sparing the differentiation of anti-inflammatory cells (Th2 and Tregs) [199].

Remarkably, despite the promising anti-GVHD potential of targeting NLRP3 and its downstream proteins, NLRP3 remains essential for HSC mobilization and homing. Current mobilization protocols still encounter a 40% non-responsiveness rate, and the side effects of mobilizing agents emphasize the urgency for gentler and more effective alternatives [200]. Similar to adjuvant function, strategies such as the combination of ATP with G-CSF or AMD3100 [201], or activating P2X4 and P2X7 receptors to elicit sufficient ATP for initiating NLRP3 activation signals [202, 203], presenting potential avenues. Additionally, the utilization of potent NLRP3 activators like Nigericin to rapidly alter the ionic environment of cells to activate NLRP3 [95], or inhibiting adenosine and its effectors (e.g., HO-1 and iNOS) to eliminate their inhibitory effects on NLRP3, are also viable strategies [87]. However, it has been found that reducing HO-1 expression increased the motility and migration of leukemia cells [204], further underlining the intricate and multifaceted nature of inflammasome regulation.

Conclusion and perspective

The multifaceted regulation of inflammasomes plays a pivotal role in innate immunity and inflammatory responses, making them promising therapeutic targets in HSCT. Inflammasomes are intricately involved in the mobilization, hematopoietic function, and complications related to transplantation, highlighting their significant potential in HSCT treatment.

However, numerous challenging issues remain to be addressed. For instance, the interplay between microbiota, their metabolites, and inflammasome signaling in GVHD warrants further exploration. In the context of chronic inflammation, sustained and recurrent activation of inflammasomes can lead to long-term tissue damage and fibrosis, highlighting the importance of analyzing their role in chronic graft-versus-host disease (cGVHD). Furthermore, immune reconstitution, infection, relapse, and human leukocyte antigen (HLA) matching are critical areas that require additional investigation, particularly concerning the unresolved challenges associated with infections. Relapse continues to be a prominent cause of HSCT failure, influenced by factors such as the regrowth of minimal residual disease, clonal evolution, and immune evasion [205]. Notably, specific NLRP3 activation features in MDSCs have been identified as potential early indicators of relapse [206]. Continued research into the mechanisms of relapse, aimed at developing clinical biomarkers, represents a promising direction. However, most current studies are limited to in vivo or in vitro experiments, with a notable lack of epidemiological cohort studies to validate findings in human populations, thereby limiting their persuasiveness.

Over the forthcoming decade, the pharmacological and genetic modulation of inflammasome activation or inhibition for preventive and therapeutic purposes are poised to become a key focus of research [189]. In the domain of HSCT research, many drugs remain experimental, with limited studies on dosage optimization, drug structure refinement, and side effects. Consequently, multicenter, large-scale, prospective, randomized controlled trials are imperative. Limited awareness of the diversity of therapeutic agents hampers the application of rich molecular scaffolds. Given the safety and broad biological activity of phytochemicals, future research may yield valuable insights into these potential drugs [207]. Moreover, the dual-targeting nature of inflammasomes must be considered seriously, particularly regarding the optimal timing and method of administration.

In summary, exploring the intricate interplay between HSCT and inflammasome activation will enhance our understanding of HSCT mechanisms and facilitate the development of improved therapeutic and management strategies.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

HSCT:

Hematopoietic stem cell transplantation

HSPCs:

Hematopoietic stem and progenitor cells

GVL:

Graft-versus-leukemia

PAMPs:

Pathogen-associated molecular patterns

DAMPs:

Danger-associated molecular patterns

IL-1β:

Interleukin-1β

IL-18:

Interleukin-18

aGVHD:

Acute graft-versus-host disease

NBD:

Nucleotide-binding domain

LRR:

Leucine-rich repeat

PYD:

Pyrin domain

CARD:

Caspase recruitment domain

BIR:

Baculovirus inhibitor of apoptosis repeat

dsDNA:

Double-stranded DNA

LPS:

Lipopolysaccharide

HMGB1:

High mobility group box 1

ATP:

Adenosine triphosphate

TLRs:

Toll-like receptors

NLRs:

NOD-like receptors

TNF-α:

Tumor necrosis factor-alpha receptor

NF-κB:

Nuclear factor kappa-light-chain-enhancer of activated B cells

K+ :

Potassium ion

Ca2+ :

Calcium ion

ROS:

Reactive oxygen species

LeTx:

Anthrax lethal toxin

PPAR-γ:

Peroxisome proliferator-activated receptor gamma

MAMPs:

Microbe-associated molecular patterns

LTA:

Lipoteichoic acid

T3SS:

Type III secretion system

cGAS:

Cyclic GMP-AMP synthase

STING:

Stimulator of interferon genes

Na+ :

Sodium ions

eATP:

Extracellular ATP

HO-1:

Heme oxygenase 1

iNOS:

Inducible nitric oxide synthase

MBL:

Mannose-binding lectin

MASPs:

Mannose-binding lectin-associated serine proteases

ComC:

Complement cascade

C3:

Complement 3

CoaC:

Coagulation cascade

MAC:

Membrane attack complex

SDF-1:

Stromal cell-derived factor 1

CXCR4:

C-X-C chemokine receptor type 4

VLA-4:

Very late antigen-4

VCAM-1:

Vascular cell adhesion molecule 1

S1P:

Sphingosine-1 phosphate

SCF:

Stem cell factor

CXCL12:

Chemokine C–X–C motif chemokine 12

PGE2:

Prostaglandin E2

SP:

Substance P

NK-A:

Neurokinin A

NK:

Natural killer

TC-PTP:

T-cell protein tyrosine phosphatase

GATA-1:

GATA binding protein 1

HIF1α:

Hypoxia-inducible factor 1-alpha

TBI:

Total body irradiation

LDH:

Lactate dehydrogenase

MHC:

Major histocompatibility complex

APCs:

Host antigen-presenting cells

Tregs:

Regulatory T cells

MDSCs:

Myeloid-derived suppressor cells

IRAK4:

Interleukin-1 receptor-associated kinase 4

MUC2:

Mucin 2

cGVHD:

Chronic graft-versus-host disease

References

  1. Little MT, Storb R. History of haematopoietic stem-cell transplantation. Nat Rev Cancer. 2002;2:231–8.

    Article  CAS  PubMed  Google Scholar 

  2. Gatti RA, Meuwissen HJ, Allen HD, Hong R, Good RA. Immunological reconstitution of sex-linked lymphopenic immunological deficiency. Lancet. 1968;2:1366–9.

    Article  CAS  PubMed  Google Scholar 

  3. Thomas ED, Storb R, Fefer A, Slichter SJ, Bryant JI, Buckner CD, et al. Aplastic anaemia treated by marrow transplantation. Lancet. 1972;1:284–9.

    Article  CAS  PubMed  Google Scholar 

  4. Niederwieser D, Baldomero H, Bazuaye N, Bupp C, Chaudhri N, Corbacioglu S, et al. One and a half million hematopoietic stem cell transplants: continuous and differential improvement in worldwide access with the use of non-identical family donors. Haematologica. 2022;107:1045–53.

    Article  PubMed  Google Scholar 

  5. Chang YJ, Pei XY, Huang XJ. Haematopoietic stem-cell transplantation in China in the era of targeted therapies: current advances, challenges, and future directions. Lancet Haematol. 2022;9:e919–29.

    Article  CAS  PubMed  Google Scholar 

  6. Li Z, Rubinstein SM, Thota R, Savani M, Brissot E, Shaw BE, et al. Immune-mediated complications after hematopoietic stem cell transplantation. Biol Blood Marrow Transpl. 2016;22:1368–75.

    Article  CAS  Google Scholar 

  7. Su L, Ji B, Hu R, Lan X, Xia C. Immune-mediated neuromuscular complications after haploidendtical hematopoietic stem cell transplantation. Chin Med J (Engl). 2014;127:2865–7.

    PubMed  Google Scholar 

  8. Liu J, Cao X. Cellular and molecular regulation of innate inflammatory responses. Cell Mol Immunol. 2016;13:711–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Próchnicki T, Latz E. Inflammasomes on the crossroads of Innate Immune Recognition and Metabolic Control. Cell Metabol. 2017;26:71–93.

    Article  Google Scholar 

  10. Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10:417–26.

    Article  CAS  PubMed  Google Scholar 

  11. Hamarsheh S, Zeiser R. NLRP3 inflammasome activation in Cancer: a double-edged Sword. Front Immunol. 2020;11:1444.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Man SM, Karki R, Kanneganti TD. Molecular mechanisms and functions of pyroptosis, inflammatory caspases and inflammasomes in infectious diseases. Immunol Rev. 2017;277:61–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ram C, Jha AK, Ghosh A, Gairola S, Syed AM, Murty US et al. Targeting NLRP3 inflammasome as a promising approach for treatment of diabetic nephropathy: preclinical evidences with therapeutic approaches. Eur J Pharmacol. 2020;885:173503.

  14. Granell M, Urbano-Ispizua Á, Pons A, Aróstegui JI, Gel B, Navarro A, et al. Common variants in NLRP2 and NLRP3 genes are strong prognostic factors for the outcome of HLA-identical sibling allogeneic stem cell transplantation. Blood. 2008;112:4337–42.

    Article  CAS  PubMed  Google Scholar 

  15. Wang L, Sharif H, Vora SM, Zheng Y, Wu H. Structures and functions of the inflammasome engine. J Allergy Clin Immunol. 2021;147:2021–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chou W-C, Jha S, Linhoff MW, Ting JPY. The NLR gene family: from discovery to present day. Nat Rev Immunol. 2023;23:635–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kobe B, Kajava AV. The leucine-rich repeat as a protein recognition motif. Curr Opin Struct Biol. 2001;11:725–32.

    Article  CAS  PubMed  Google Scholar 

  18. Dixon CL, Wu A, Fairn GD. Multifaceted roles and regulation of nucleotide-binding oligomerization domain containing proteins. Front Immunol. 2023;14:1242659.

  19. Kapoor P, Briggs DA, Cox MH, Kerr ID. Disruption of the unique ABCG-family NBD:NBD interface impacts both drug transport and ATP hydrolysis. Int J Mol Sci. 2020;21:759.

  20. Finger JN, Lich JD, Dare LC, Cook MN, Brown KK, Duraiswami C, et al. Autolytic Proteolysis within the function to find domain (FIIND) is required for NLRP1 inflammasome activity. J Biol Chem. 2012;287:25030–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Martín-Sánchez F, Compan V, Peñín-Franch A, Tapia-Abellán A, Gómez AI, Baños-Gregori MC et al. ASC oligomer favors caspase-1CARD domain recruitment after intracellular potassium efflux. J Cell Biol. 2023;222:e202003053.

  22. Jin T, Perry A, Jiang J, Smith P, Curry JA, Unterholzner L, et al. Structures of the HIN domain:DNA complexes reveal ligand binding and activation mechanisms of the AIM2 inflammasome and IFI16 receptor. Immunity. 2012;36:561–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wang B, Tian Y, Yin Q. AIM2 Inflammasome Assembly and Signaling. Adv Exp Med Biol. 2019;1172:143–55.

    Article  CAS  PubMed  Google Scholar 

  24. Wang B, Yin Q. AIM2 inflammasome activation and regulation: a structural perspective. J Struct Biol. 2017;200:279–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature. 2009;458:514–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Veeranki S, Choubey D. Interferon-inducible p200-family protein IFI16, an innate immune sensor for cytosolic and nuclear double-stranded DNA: regulation of subcellular localization. Mol Immunol. 2012;49:567–71.

    Article  CAS  PubMed  Google Scholar 

  27. Schnappauf O, Chae JJ, Kastner DL, Aksentijevich I. The pyrin inflammasome in health and disease. Front Immunol. 2019;10:1745.

  28. Chae JJ, Wood G, Masters SL, Richard K, Park G, Smith BJ, et al. The B30.2 domain of pyrin, the familial Mediterranean fever protein, interacts directly with caspase-1 to modulate IL-1beta production. Proc Natl Acad Sci U S A. 2006;103:9982–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhai Y, Wang C, Jiang Z. Cross-talk between bacterial PAMPs and host PRRs. Natl Sci Rev. 2018;5:791–2.

    Article  CAS  Google Scholar 

  30. Ma M, Jiang W, Zhou R. DAMPs and DAMP-sensing receptors in inflammation and diseases. Immunity. 2024;57:752–71.

    Article  CAS  PubMed  Google Scholar 

  31. Chen H, Jiang Z. The essential adaptors of innate immune signaling. Protein Cell. 2012;4:27–39.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Fukata M, Vamadevan AS, Abreu MT. Toll-like receptors (TLRs) and nod-like receptors (NLRs) in inflammatory disorders. Semin Immunol. 2009;21:242–53.

    Article  CAS  PubMed  Google Scholar 

  33. Yang J, Liu Z, Xiao TS. Post-translational regulation of inflammasomes. Cell Mol Immunol. 2016;14:65–79.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Akbal A, Dernst A, Lovotti M, Mangan MSJ, McManus RM, Latz E. How location and cellular signaling combine to activate the NLRP3 inflammasome. Cell Mol Immunol. 2022;19:1201–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Young JAT, Levinsohn JL, Newman ZL, Hellmich KA, Fattah R, Getz MA et al. Anthrax Lethal factor cleavage of Nlrp1 is required for activation of the Inflammasome. PLoS Pathog. 2012;8:e1002638.

  36. Bauernfried S, Scherr MJ, Pichlmair A, Duderstadt KE, Hornung V. Human NLRP1 is a sensor for double-stranded RNA. Science. 2021;371:eabd0811.

  37. Tsu BV, Beierschmitt C, Ryan AP, Agarwal R, Mitchell PS, Daugherty MD. Diverse viral proteases activate the NLRP1 inflammasome. Elife. 2021;10:e60609.

  38. Jenster L-M, Lange K-E, Normann S, vom Hemdt A, Wuerth JD, Schiffelers LDJ et al. P38 kinases mediate NLRP1 inflammasome activation after ribotoxic stress response and virus infection. J Exp Med. 2023;220:e20220837.

  39. Muela-Zarzuela I, Suarez-Rivero JM, Gallardo-Orihuela A, Wang C, Izawa K, de Gregorio-Procopio M, et al. NLRP1 inflammasome promotes senescence and senescence-associated secretory phenotype. Inflamm Res. 2024;73:1253–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wang P, Zhu S, Yang L, Cui S, Pan W, Jackson R, et al. Nlrp6 regulates intestinal antiviral innate immunity. Science. 2015;350:826–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kempster SL, Belteki G, Forhead AJ, Fowden AL, Catalano RD, Lam BY, et al. Developmental control of the Nlrp6 inflammasome and a substrate, IL-18, in mammalian intestine. Am J Physiol Gastrointest Liver Physiol. 2011;300:G253–63.

    Article  CAS  PubMed  Google Scholar 

  42. Li R, Zhu S. NLRP6 inflammasome. Mol Aspects Med. 2020;76:100859.

    Article  CAS  PubMed  Google Scholar 

  43. Hara H, Seregin SS, Yang D, Fukase K, Chamaillard M, Alnemri ES, et al. The NLRP6 Inflammasome recognizes Lipoteichoic Acid and regulates gram-positive Pathogen infection. Cell. 2018;175:1651–e6414.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Leng F, Yin H, Qin S, Zhang K, Guan Y, Fang R, et al. NLRP6 self-assembles into a linear molecular platform following LPS binding and ATP stimulation. Sci Rep. 2020;10:198.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Levy M, Thaiss Christoph A, Zeevi D, Dohnalová L, Zilberman-Schapira G, Mahdi Jemal A, et al. Microbiota-modulated metabolites shape the Intestinal Microenvironment by regulating NLRP6 Inflammasome Signaling. Cell. 2015;163:1428–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Zhao Y, Yang J, Shi J, Gong YN, Lu Q, Xu H, et al. The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus. Nature. 2011;477:596–600.

    Article  CAS  PubMed  Google Scholar 

  47. Morrone SR, Matyszewski M, Yu X, Delannoy M, Egelman EH, Sohn J. Assembly-driven activation of the AIM2 foreign-dsDNA sensor provides a polymerization template for downstream ASC. Nat Commun. 2015;6:7827.

  48. Rathinam VAK, Jiang Z, Waggoner SN, Sharma S, Cole LE, Waggoner L, et al. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat Immunol. 2010;11:395–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Zhu Q, Man SM, Karki R, Malireddi RKS, Kanneganti T-D. Detrimental type I Interferon Signaling dominates protective AIM2 inflammasome responses during Francisella novicida infection. Cell Rep. 2018;22:3168–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339:786–91.

    Article  CAS  PubMed  Google Scholar 

  51. Almine JF, O’Hare CA, Dunphy G, Haga IR, Naik RJ, Atrih A, et al. IFI16 and cGAS cooperate in the activation of STING during DNA sensing in human keratinocytes. Nat Commun. 2017;8:14392.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Huang Y, Xu W, Zhou R. NLRP3 inflammasome activation and cell death. Cell Mol Immunol. 2021;18:2114–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yu T-G, Cha JS, Kim G, Sohn Y-K, Yoo Y, Kim U et al. Oligomeric states of ASC specks regulate inflammatory responses by inflammasome in the extracellular space. Cell Death Discovery. 2023;9:142.

  54. Dick MS, Sborgi L, Rühl S, Hiller S, Broz P. ASC filament formation serves as a signal amplification mechanism for inflammasomes. Nat Commun. 2016;7:11929.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. de Alba E. Structure, interactions and self-assembly of ASC-dependent inflammasomes. Arch Biochem Biophys. 2019;670:15–31.

    Article  PubMed  PubMed Central  Google Scholar 

  56. Boucher D, Monteleone M, Coll RC, Chen KW, Ross CM, Teo JL, et al. Caspase-1 self-cleavage is an intrinsic mechanism to terminate inflammasome activity. J Exp Med. 2018;215:827–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yu HB, Finlay BB. The caspase-1 inflammasome: a pilot of innate immune responses. Cell Host Microbe. 2008;4:198–208.

    Article  CAS  PubMed  Google Scholar 

  58. Kesavardhana S, Malireddi RKS, Kanneganti T-D. Caspases in cell death, inflammation, and Pyroptosis. Annu Rev Immunol. 2020;38:567–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7:99–109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Broz P, Pelegrín P, Shao F. The gasdermins, a protein family executing cell death and inflammation. Nat Rev Immunol. 2020;20:143–57.

    Article  CAS  PubMed  Google Scholar 

  61. Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, et al. Pore-forming activity and structural autoinhibition of the gasdermin family. Nature. 2016;535:111–6.

    Article  CAS  PubMed  Google Scholar 

  62. Devant P, Kagan JC. Molecular mechanisms of gasdermin D pore-forming activity. Nat Immunol. 2023;24:1064–75.

    Article  CAS  PubMed  Google Scholar 

  63. Shi J, Zhao Y, Wang Y, Gao W, Ding J, Li P, et al. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature. 2014;514:187–92.

    Article  CAS  PubMed  Google Scholar 

  64. Baker PJ, Boucher D, Bierschenk D, Tebartz C, Whitney PG, D’Silva DB, et al. NLRP3 inflammasome activation downstream of cytoplasmic LPS recognition by both caspase-4 and caspase-5. Eur J Immunol. 2015;45:2918–26.

    Article  CAS  PubMed  Google Scholar 

  65. Fritsch M, Günther SD, Schwarzer R, Albert MC, Schorn F, Werthenbach JP, et al. Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature. 2019;575:683–7.

    Article  CAS  PubMed  Google Scholar 

  66. Sagulenko V, Vitak N, Vajjhala PR, Vince JE, Stacey KJ. Caspase-1 is an apical caspase leading to Caspase-3 cleavage in the AIM2 Inflammasome Response, Independent of Caspase-8. J Mol Biol. 2018;430:238–47.

    Article  CAS  PubMed  Google Scholar 

  67. Oh S, Lee J, Oh J, Yu G, Ryu H, Kim D, et al. Integrated NLRP3, AIM2, NLRC4, pyrin inflammasome activation and assembly drive PANoptosis. Cell Mol Immunol. 2023;20:1513–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Xu Z, Kombe Kombe AJ, Deng S, Zhang H, Wu S, Ruan J, et al. NLRP inflammasomes in health and disease. Mol Biomed. 2024;5:14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zindel J, Kubes P. DAMPs, PAMPs, and LAMPs in immunity and sterile inflammation. Annu Rev Pathol. 2020;15:493–518.

    Article  CAS  PubMed  Google Scholar 

  70. Patel S. Inflammasomes, the cardinal pathology mediators are activated by pathogens, allergens and mutagens: a critical review with focus on NLRP3. Biomed Pharmacother. 2017;92:819–25.

    Article  CAS  PubMed  Google Scholar 

  71. Christgen S, Kanneganti TD. Inflammasomes and the fine line between defense and disease. Curr Opin Immunol. 2020;62:39–44.

    Article  CAS  PubMed  Google Scholar 

  72. Brewer SM, Brubaker SW, Monack DM. Host inflammasome defense mechanisms and bacterial pathogen evasion strategies. Curr Opin Immunol. 2019;60:63–70.

    Article  CAS  PubMed  Google Scholar 

  73. Li Y, Huang H, Liu B, Zhang Y, Pan X, Yu XY, et al. Inflammasomes as therapeutic targets in human diseases. Signal Transduct Target Ther. 2021;6:247.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Shadab A, Mahjoor M, Abbasi-Kolli M, Afkhami H, Moeinian P, Safdarian AR. Divergent functions of NLRP3 inflammasomes in cancer: a review. Cell Commun Signal. 2023;21:232.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Karki R, Man SM, Kanneganti TD. Inflammasomes and Cancer. Cancer Immunol Res. 2017;5:94–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Mishra SR, Mahapatra KK, Behera BP, Bhol CS, Praharaj PP, Panigrahi DP, et al. Inflammasomes in cancer: Effect of epigenetic and autophagic modulations. Semin Cancer Biol. 2022;83:399–412.

    Article  CAS  PubMed  Google Scholar 

  77. Whetton AD, Graham GJ. Homing and mobilization in the stem cell niche. Trends Cell Biol. 1999;9:233–8.

    Article  CAS  PubMed  Google Scholar 

  78. Christopher MJ, Rao M, Liu F, Woloszynek JR, Link DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J Exp Med. 2011;208:251–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Velders GA, van Os R, Hagoort H, Verzaal P, Guiot HF, Lindley IJ, et al. Reduced stem cell mobilization in mice receiving antibiotic modulation of the intestinal flora: involvement of endotoxins as cofactors in mobilization. Blood. 2004;103:340–6.

    Article  CAS  PubMed  Google Scholar 

  80. Schuettpelz LG, Borgerding JN, Christopher MJ, Gopalan PK, Romine MP, Herman AC, et al. G-CSF regulates hematopoietic stem cell activity, in part, through activation of toll-like receptor signaling. Leukemia. 2014;28:1851–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Narahari AK, Kreutzberger AJ, Gaete PS, Chiu YH, Leonhardt SA, Medina CB et al. ATP and large signaling metabolites flux through caspase-activated Pannexin 1 channels. Elife. 2021;10:e64787.

  82. Cymer M, Brzezniakiewicz-Janus K, Bujko K, Thapa A, Ratajczak J, Anusz K, et al. Pannexin-1 channel fuels by releasing ATP from bone marrow cells a state of sterile inflammation required for optimal mobilization and homing of hematopoietic stem cells. Purinergic Signal. 2020;16:313–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Vergani A, Tezza S, Fotino C, Visner G, Pileggi A, Chandraker A, et al. The purinergic system in allotransplantation. Am J Transpl. 2014;14:507–14.

    Article  CAS  Google Scholar 

  84. Adamiak M, Bujko K, Thapa A, Pensato V, Brzezniakiewicz-Janus K, Ratajczak J, et al. The P2X4 purinergic receptor has emerged as a potent regulator of hematopoietic stem/progenitor cell mobilization and homing-a novel view of P2X4 and P2X7 receptor interaction in orchestrating stem cell trafficking. Leukemia. 2022;36:248–56.

    Article  CAS  PubMed  Google Scholar 

  85. Bujko K, Adamiak M, Abdelbaset-Ismail A, Thapa A, Ilowska N, Ratajczak J, et al. Novel evidence that the P2X1 purinergic receptor-Nlrp3 inflammasome axis orchestrates optimal trafficking of hematopoietic stem progenitors cells. Folia Histochem Cytobiol. 2022;60:280–90.

    Article  CAS  PubMed  Google Scholar 

  86. Jiang X, Wu X, Xiao Y, Wang P, Zheng J, Wu X, et al. The ectonucleotidases CD39 and CD73 on T cells: the new pillar of hematological malignancy. Front Immunol. 2023;14:1110325.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Ratajczak MZ, Adamiak M, Ratajczak J, Kucia M. Heme Oxygenase 1 (HO-1) as an inhibitor of trafficking of normal and malignant hematopoietic stem cells - clinical and translational implications. Stem Cell Rev Rep. 2021;17:821–8.

    Article  CAS  PubMed  Google Scholar 

  88. Adamiak M, Abdelbaset-Ismail A, Moore JBt, Zhao J, Abdel-Latif A, Wysoczynski M, et al. Inducible nitric oxide synthase (iNOS) is a novel negative Regulator of hematopoietic Stem/Progenitor cell trafficking. Stem Cell Rev Rep. 2017;13:92–103.

    Article  CAS  PubMed  Google Scholar 

  89. Thapa A, Abdelbaset-Ismail A, Chumak V, Adamiak M, Brzezniakiewicz-Janus K, Ratajczak J, et al. Extracellular Adenosine (eAdo) - A(2B) receptor Axis inhibits in Nlrp3 inflammasome-dependent manner trafficking of hematopoietic Stem/progenitor cells. Stem Cell Rev Rep. 2022;18:2893–911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Ratajczak MZ, Adamiak M, Thapa A, Bujko K, Brzezniakiewicz-Janus K, Lenkiewicz AM. NLRP3 inflammasome couples purinergic signaling with activation of the complement cascade for the optimal release of cells from bone marrow. Leukemia. 2019;33:815–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Cymer M, Brzezniakiewicz-Janus K, Bujko K, Thapa A, Ratajczak J, Anusz K, et al. Pannexin-1 channel fuels by releasing ATP from bone marrow cells a state of sterile inflammation required for optimal mobilization and homing of hematopoietic stem cells. Purinergic Signalling. 2020;16:313–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Thapa A, Adamiak M, Bujko K, Ratajczak J, Abdel-Latif AK, Kucia M, et al. Danger-associated molecular pattern molecules take unexpectedly a central stage in Nlrp3 inflammasome–caspase-1-mediated trafficking of hematopoietic stem/progenitor cells. Leukemia. 2021;35:2658–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Adamiak M, Abdelbaset-Ismail A, Suszynska M, Abdel-Latif A, Ratajczak J, Ratajczak MZ. Novel evidence that the mannan-binding lectin pathway of complement activation plays a pivotal role in triggering mobilization of hematopoietic stem/progenitor cells by activation of both the complement and coagulation cascades. Leukemia. 2016;31:262–5.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Adamiak M, Cymer M, Anusz K, Tracz M, Ratajczak MZ. A Novel evidence that Mannan binding lectin (MBL) pathway of Complement Cascade activation is involved in Homing and Engraftment of hematopoietic stem progenitor cells (HSPCs). Stem Cell Reviews Rep. 2020;16:693–701.

    Article  CAS  Google Scholar 

  95. Lenkiewicz AM, Adamiak M, Thapa A, Bujko K, Pedziwiatr D, Abdel-Latif AK, et al. The Nlrp3 Inflammasome orchestrates mobilization of bone marrow-residing stem cells into peripheral blood. Stem Cell Rev Rep. 2019;15:391–403.

    Article  CAS  PubMed  Google Scholar 

  96. Adamiak M, Ciechanowicz A, Chumak V, Bujko K, Ratajczak J, Brzezniakiewicz-Janus K, et al. Novel evidence that Alternative Pathway of Complement Cascade activation is required for optimal homing and engraftment of hematopoietic Stem/progenitor cells. Stem Cell Reviews Rep. 2022;18:1355–65.

    Article  CAS  Google Scholar 

  97. Ratajczak MZ, Adamiak M, Abdelbaset-Ismail A, Bujko K, Thapa A, Chumak V, et al. Intracellular complement (complosome) is expressed in hematopoietic stem/progenitor cells (HSPCs) and regulates cell trafficking, metabolism and proliferation in an intracrine Nlrp3 inflammasome-dependent manner. Leukemia. 2023;37:1401–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Borkowska S, Suszynska M, Mierzejewska K, Ismail A, Budkowska M, Salata D, et al. Novel evidence that crosstalk between the complement, coagulation and fibrinolysis proteolytic cascades is involved in mobilization of hematopoietic stem/progenitor cells (HSPCs). Leukemia. 2014;28:2148–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Petit I, Szyper-Kravitz M, Nagler A, Lahav M, Peled A, Habler L, et al. G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4. Nat Immunol. 2002;3:687–94.

    Article  CAS  PubMed  Google Scholar 

  100. Vermeulen M, Le Pesteur F, Gagnerault MC, Mary JY, Sainteny F, Lepault F. Role of adhesion molecules in the homing and mobilization of murine hematopoietic stem and progenitor cells. Blood. 1998;92:894–900.

    Article  CAS  PubMed  Google Scholar 

  101. Rettig MP, Ansstas G, DiPersio JF. Mobilization of hematopoietic stem and progenitor cells using inhibitors of CXCR4 and VLA-4. Leukemia. 2011;26:34–53.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Lee HM, Wu W, Wysoczynski M, Liu R, Zuba-Surma EK, Kucia M, et al. Impaired mobilization of hematopoietic stem/progenitor cells in C5-deficient mice supports the pivotal involvement of innate immunity in this process and reveals novel promobilization effects of granulocytes. Leukemia. 2009;23:2052–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Ratajczak MZ, Adamiak M, Plonka M, Abdel-Latif A, Ratajczak J. Mobilization of hematopoietic stem cells as a result of innate immunity-mediated sterile inflammation in the bone marrow microenvironment—the involvement of extracellular nucleotides and purinergic signaling. Leukemia. 2018;32:1116–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Ratajczak MZ, Lee H, Wysoczynski M, Wan W, Marlicz W, Laughlin MJ, et al. Novel insight into stem cell mobilization-plasma sphingosine-1-phosphate is a major chemoattractant that directs the egress of hematopoietic stem progenitor cells from the bone marrow and its level in peripheral blood increases during mobilization due to activation of complement cascade/membrane attack complex. Leukemia. 2010;24:976–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Ratajczak J, Reca R, Kucia M, Majka M, Allendorf DJ, Baran JT, et al. Mobilization studies in mice deficient in either C3 or C3a receptor (C3aR) reveal a novel role for complement in retention of hematopoietic stem/progenitor cells in bone marrow. Blood. 2004;103:2071–8.

    Article  CAS  PubMed  Google Scholar 

  106. Ratajczak MZ, Adamiak M. Membrane lipid rafts, master regulators of hematopoietic stem cell retention in bone marrow and their trafficking. Leukemia. 2015;29:1452–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Abdelbaset-Ismail A, Ciechanowicz AK, Bujko K, Ratajczak J, Kucia M, Ratajczak MZ. The Nox2-ROS-Nlrp3 Inflammasome Signaling stimulates in the hematopoietic Stem/Progenitor cells lipogenesis to facilitate membrane lipid raft formation. Stem Cell Reviews Rep. 2022;19:92–103.

    Article  Google Scholar 

  108. Adamiak M, Abdel-Latif A, Bujko K, Thapa A, Anusz K, Tracz M, et al. Nlrp3 Inflammasome Signaling regulates the Homing and Engraftment of hematopoietic stem cells (HSPCs) by enhancing incorporation of CXCR4 receptor into membrane lipid rafts. Stem Cell Reviews Rep. 2020;16:954–67.

    Article  CAS  Google Scholar 

  109. Park B, Yoo KH, Kim C. Hematopoietic stem cell expansion and generation: the ways to make a breakthrough. Blood Res. 2015;50:194–203.

  110. Man Y, Lu Z, Yao X, Gong Y, Yang T, Wang Y. Recent advancements in poor graft function following hematopoietic stem cell transplantation. Front Immunol. 2022;13:911174.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Pinho S, Frenette PS. Haematopoietic stem cell activity and interactions with the niche. Nat Rev Mol Cell Biol. 2019;20:303–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Wu J, Zhang W, Ran Q, Xiang Y, Zhong JF, Li SC, et al. The differentiation balance of bone marrow mesenchymal stem cells is crucial to Hematopoiesis. Stem Cells Int. 2018;2018:1–13.

    CAS  Google Scholar 

  113. Hong F, Chen Y, Gao H, Shi J, Lu W, Ju W et al. NLRP1 in Bone Marrow Microenvironment Controls Hematopoietic Reconstitution After Transplantation. Transplantation and Cellular Therapy. 2021;27:908.e1-.e11.

  114. Ambrosi TH, Scialdone A, Graja A, Gohlke S, Jank AM, Bocian C et al. Adipocyte Accumulation in the bone marrow during obesity and aging impairs stem cell-based hematopoietic and bone regeneration. Cell Stem Cell. 2017;20:771–84.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Zhang J, Chen Y, Yin D, Feng F, An Q, Liu Z et al. Caspase-3/NLRP3 signaling in the mesenchymal stromal niche regulates myeloid-biased hematopoiesis. Stem Cell Res Ther. 2021;12:579.

  116. Park HS, Lee B-C, Chae D-H, Yu A, Park JH, Heo J et al. Cigarette smoke impairs the hematopoietic supportive property of mesenchymal stem cells via the production of reactive oxygen species and NLRP3 activation. Stem Cell Res Ther. 2024;15:145.

  117. Liu K, Castillo MD, Murthy RG, Patel N, Rameshwar P. Tachykinins and Hematopoiesis. Clin Chim Acta. 2007;385:28–34.

    Article  CAS  PubMed  Google Scholar 

  118. Gergues M, Nagula V, Bliss SA, Eljarrah A, Ayer S, Gnanavel N, et al. Neuroimmune/Hematopoietic Axis with distinct regulation by the High-Mobility Group Box 1 in Association with tachykinin peptides. J Immunol. 2020;204:879–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Wilkinson AC, Igarashi KJ, Nakauchi H. Haematopoietic stem cell self-renewal in vivo and ex vivo. Nat Rev Genet. 2020;21:541–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Bourdeau A, Dubé N, Tremblay ML. Cytoplasmic protein tyrosine phosphatases, regulation and function: the roles of PTP1B and TC-PTP. Curr Opin Cell Biol. 2005;17:203–9.

    Article  CAS  PubMed  Google Scholar 

  121. Bourdeau A, Trop S, Doody KM, Dumont DJ, Tremblayef ML. Inhibition of T cell protein tyrosine phosphatase enhances Interleukin-18-Dependent hematopoietic stem cell expansion. Stem Cells. 2013;31:293–304.

    Article  CAS  PubMed  Google Scholar 

  122. Radujkovic A, Kordelas L, Dai H, Schult D, Majer-Lauterbach J, Beelen D, et al. Interleukin-18 and outcome after allogeneic stem cell transplantation: a retrospective cohort study. eBioMedicine. 2019;49:202–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Tyrkalska SD, Pérez-Oliva AB, Rodríguez-Ruiz L, Martínez-Morcillo FJ, Alcaraz-Pérez F, Martínez-Navarro FJ, et al. Inflammasome regulates hematopoiesis through cleavage of the master erythroid transcription factor GATA1. Immunity. 2019;51:50–e635.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. De Maria R, Zeuner A, Eramo A, Domenichelli C, Bonci D, Grignani F, et al. Negative regulation of erythropoiesis by caspase-mediated cleavage of GATA-1. Nature. 1999;401:489–93.

    Article  PubMed  Google Scholar 

  125. Rodríguez-Ruiz L, Lozano‐Gil JM, Naranjo‐Sánchez E, Martínez‐Balsalobre E, Martínez‐López A, Lachaud C et al. ZAKα/P38 kinase signaling pathway regulates hematopoiesis by activating the NLRP1 inflammasome. EMBO Mol Med. 2023;15:e18142.

  126. Frame J, Long T, Schuster-Kubaczka C, Esain V, Lim SE, Daley G et al. Inflammasome-mediated expansion of the hematopoietic system in the Vertebrate embryo. Exp Hematol. 2019;76:S52–96.

  127. Frame JM, Kubaczka C, Long TL, Esain V, Soto RA, Hachimi M et al. Metabolic regulation of Inflammasome Activity controls Embryonic hematopoietic stem and progenitor cell production. Dev Cell. 2020;55:133–49.e6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Qiao J, Wu J, Li Y, Xia Y, Chu P, Qi K, et al. Blockage of caspase-1 activation ameliorates bone marrow inflammation in mice after hematopoietic stem cell transplantation. Clin Immunol. 2016;162:84–90.

    Article  CAS  PubMed  Google Scholar 

  129. Hu B, Jin C, Li HB, Tong J, Ouyang X, Cetinbas NM, et al. The DNA-sensing AIM2 inflammasome controls radiation-induced cell death and tissue injury. Science. 2016;354:765–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Liu Y-g, Chen J-k, Zhang Z-t, Ma X-j, Chen Y-c, Du X-m, et al. NLRP3 inflammasome activation mediates radiation-induced pyroptosis in bone marrow-derived macrophages. Cell Death Dis. 2017;8:e2579–e.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Linz BML, Neely CJ, Kartchner LB, Mendoza AE, Khoury AL, Truax A, et al. Innate Immune Cell Recovery is positively regulated by NLRP12 during Emergency Hematopoiesis. J Immunol. 2017;198:2426–33.

    Article  CAS  PubMed  Google Scholar 

  132. Yang X, Cong T, He H, Wang J. GSDME maintains hematopoietic stem cells by balancing pyroptosis and apoptosis. Blood Sci. 2021;3:40–7.

    Article  PubMed  Google Scholar 

  133. Patel DA, Crain M, Pusic I, Schroeder MA. Acute Graft-versus-host disease: an update on New Treatment options. Drugs. 2023;83:893–907.

    Article  PubMed  Google Scholar 

  134. Malard F, Holler E, Sandmaier BM, Huang H, Mohty M. Acute graft-versus-host disease. Nat Reviews Disease Primers. 2023;9:27.

  135. Hill GR, Koyama M. Cytokines and costimulation in acute graft-versus-host disease. Blood. 2020;136:418–28.

    Article  PubMed  PubMed Central  Google Scholar 

  136. Al-Hashmi S, Hassan Z, Sadeghi B, Rozell B, Hassan M. Dynamics of early histopathological changes in GVHD after busulphan/cyclophosphamide conditioning regimen. Int J Clin Exp Pathol. 2011;4:596–605.

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Toubai T, Mathewson ND, Magenau J, Reddy P. Danger signals and graft-versus-host disease: current understanding and future perspectives. Front Immunol. 2016;7:539.

  138. Wysocki CA, Panoskaltsis-Mortari A, Blazar BR, Serody JS. Leukocyte migration and graft-versus-host disease. Blood. 2005;105:4191–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Hwang JR, Byeon Y, Kim D, Park SG. Recent insights of T cell receptor-mediated signaling pathways for T cell activation and development. Exp Mol Med. 2020;52:750–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Campe J, Ullrich E. T helper cell lineage-defining transcription factors: potent targets for specific GVHD therapy? Front Immunol. 2022;12:806529.

  141. Du W, Cao X. Cytotoxic pathways in allogeneic hematopoietic cell transplantation. Front Immunol. 2018;9:2979.

  142. Koehn BH, Saha A, McDonald-Hyman C, Loschi M, Thangavelu G, Ma L, et al. Danger-associated extracellular ATP counters MDSC therapeutic efficacy in acute GVHD. Blood. 2019;134:1670–82.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Zhu S, Shi P, Lv C, Li H, Pan B, Chen W, et al. Loss of NLRP3 function alleviates murine hepatic graft-versus-host disease. Biol Blood Marrow Transplant. 2018;24:2409–17.

    Article  CAS  PubMed  Google Scholar 

  144. Koldej RM, Perera T, Collins J, Ritchie DS. Association between P2X7 polymorphisms and post-transplant outcomes in allogeneic haematopoietic stem cell transplantation. Int J Mol Sci. 2020;21:3772.

  145. Jankovic D, Ganesan J, Bscheider M, Stickel N, Weber FC, Guarda G, et al. The Nlrp3 inflammasome regulates acute graft-versus-host disease. J Exp Med. 2013;210:1899–910.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Chiusolo P, Giammarco S, Fanali C, Bellesi S, Metafuni E, Sica S, et al. Salivary proteomic analysis and Acute Graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2013;19:888–92.

    Article  CAS  PubMed  Google Scholar 

  147. Young JW. Alternative mechanisms that mediate graft-versus-host disease in allogeneic hematopoietic cell transplants. J Clin Invest. 2020;130:4532–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Chen S, Smith BAH, Iype J, Prestipino A, Pfeifer D, Grundmann S, et al. MicroRNA-155–deficient dendritic cells cause less severe GVHD through reduced migration and defective inflammasome activation. Blood. 2015;126:103–12.

    Article  CAS  PubMed  Google Scholar 

  149. Lu Y, Meng R, Wang X, Xu Y, Tang Y, Wu J et al. Caspase-11 signaling enhances graft-versus-host disease. Nat Commun. 2019;10:4044.

  150. Yujiri T, Tagami K, Tanaka Y, Mitani N, Nakamura Y, Ariyoshi K, et al. Increased serum levels of high-mobility group box 1 protein in patients who developed acute graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Eur J Haematol. 2010;85:366–7.

    Article  PubMed  Google Scholar 

  151. Xu YJ, Li L, Chen Y, Fu B, Wu DS, Li XL, et al. Role of HMGB1 in regulation of STAT3 expression in CD4(+) T cells from patients with aGVHD after allogeneic hematopoietic stem cell transplantation. Clin Immunol. 2015;161:278–83.

    Article  CAS  PubMed  Google Scholar 

  152. Wu K, Yuan Y, Yu H, Dai X, Wang S, Sun Z, et al. The gut microbial metabolite trimethylamine N-oxide aggravates GVHD by inducing M1 macrophage polarization in mice. Blood. 2020;136:501–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Roy S, Rizvi ZA, Awasthi A. Metabolic checkpoints in differentiation of helper T cells in tissue inflammation. Front Immunol. 2018;9:3036.

    Article  CAS  PubMed  Google Scholar 

  154. Talley S, Rademacher DJ, Campbell EM. Inflammasome activation occurs in CD4 + and CD8 + T cells during graft-versus-host disease. Cell Death Dis. 2023;14:632.

  155. Cheong M, Gartlan KH, Lee JS, Tey S-K, Zhang P, Kuns RD, et al. ASC modulates CTL cytotoxicity and transplant outcome Independent of the Inflammasome. Cancer Immunol Res. 2020;8:1085–98.

    Article  CAS  PubMed  Google Scholar 

  156. Yao Y, Chen S, Cao M, Fan X, Yang T, Huang Y et al. Antigen-specific CD8 + T cell feedback activates NLRP3 inflammasome in antigen-presenting cells through perforin. Nat Commun. 2017;8:15402.

  157. Shaiegan M, Iravani M, Babaee GR, Ghavamzadeh A. Effect of IL-18 and sIL2R on aGVHD occurrence after hematopoietic stem cell transplantation in some Iranian patients. Transpl Immunol. 2006;15:223–7.

    Article  CAS  PubMed  Google Scholar 

  158. Li X, Zhang C, Chen WEI, Pan BIN, Kong F, Zheng K, et al. Protective effect of neutralizing anti-IL-18α monoclonal antibody on a mouse model of acute graft-versus-host disease. Oncol Rep. 2015;34:2031–9.

    Article  CAS  PubMed  Google Scholar 

  159. Godfrey DI, Koay HF, McCluskey J, Gherardin NA. The biology and functional importance of MAIT cells. Nat Immunol. 2019;20:1110–28.

    Article  CAS  PubMed  Google Scholar 

  160. Gao M-G, Hong Y, Zhao X-Y, Pan X-A, Sun Y-Q, Kong J et al. The potential roles of mucosa-associated Invariant T Cells in the pathogenesis of gut graft-versus-host disease after hematopoietic stem cell transplantation. Front Immunol. 2021;12:720354.

  161. Naymagon S, Naymagon L, Wong SY, Ko HM, Renteria A, Levine J, et al. Acute graft-versus-host disease of the gut: considerations for the gastroenterologist. Nat Rev Gastroenterol Hepatol. 2017;14:711–26.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Au-Yeung BB, Shah NH, Shen L, Weiss A. ZAP-70 in Signaling, Biology, and Disease. Annu Rev Immunol. 2018;36:127–56.

    Article  CAS  PubMed  Google Scholar 

  163. Suto M, Matsuki E, Miyata M, Sekiguchi E, Tamaki H, Tawara I, et al. The absence of NLRP6 in Donor T cells exacerbates Gvhd. Blood. 2021;138:2766.

    Article  Google Scholar 

  164. Zhu S, Zhang X, Xu K, Liang J, Wang W, Zeng L, et al. Loss of NLRP6 expression increases the severity of intestinal injury after syngeneic hematopoietic stem cell transplantation. Ann Hematol. 2024;103:3145–54.

    Article  CAS  PubMed  Google Scholar 

  165. Li M, Chen Y, Shi J, Ju W, Qi K, Fu C et al. NLRP6 deficiency aggravates liver injury after allogeneic hematopoietic stem cell transplantation. Int Immunopharmacol. 2019;74:105740.

  166. Toubai T, Fujiwara H, Rossi C, Riwes M, Tamaki H, Zajac C, et al. Host NLRP6 exacerbates graft-versus-host disease independent of gut microbial composition. Nat Microbiol. 2019;4:800–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Toubai T, Rossi C, Oravecz-Wilson K, Tamaki H, Liu C, Mathewson N, et al. NLRP6 in host target tissues exacerbates graft-versus-host-disease. Biol Blood Marrow Transplant. 2016;22:S415–6.

    Article  Google Scholar 

  168. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Fujiwara H, Docampo MD, Riwes M, Peltier D, Toubai T, Henig I et al. Microbial metabolite sensor GPR43 controls severity of experimental GVHD. Nat Commun. 2018;9:3674.

  170. Ghimire S, Weber D, Hippe K, Meedt E, Hoepting M, Kattner A-S et al. GPR expression in intestinal biopsies from SCT patients is upregulated in GvHD and is suppressed by broad-spectrum antibiotics. Front Immunol. 2021;12:753287.

  171. Styczyński J, Tridello G, Koster L, Iacobelli S, van Biezen A, van der Werf S, et al. Death after hematopoietic stem cell transplantation: changes over calendar year time, infections and associated factors. Bone Marrow Transplant. 2019;55:126–36.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Sutrave G, Blyth E, Gottlieb DJ. Cellular therapy for multiple pathogen infections after hematopoietic stem cell transplant. Cytotherapy. 2017;19:1284–301.

    Article  CAS  PubMed  Google Scholar 

  173. Wingard JR, Hsu J, Hiemenz JW. Hematopoietic stem cell transplantation: an overview of infection risks and epidemiology. Infect Dis Clin N Am. 2010;24:257–72.

    Article  Google Scholar 

  174. Sahin U, Toprak SK, Atilla PA, Atilla E, Demirer T. An overview of infectious complications after allogeneic hematopoietic stem cell transplantation. J Infect Chemother. 2016;22:505–14.

    Article  PubMed  Google Scholar 

  175. Huang Y, Liu L, Ma D, Liao Y, Lu Y, Huang H, et al. Human cytomegalovirus triggers the assembly of AIM2 inflammasome in THP-1-derived macrophages. J Med Virol. 2017;89:2188–95.

    Article  CAS  PubMed  Google Scholar 

  176. Coulon P-G, Dhanushkodi N, Prakash S, Srivastava R, Roy S, Alomari NI et al. NLRP3, NLRP12, and IFI16 inflammasomes induction and caspase-1 activation triggered by virulent HSV-1 strains are associated with severe corneal inflammatory herpetic disease. Front Immunol. 2019;10:1631.

  177. Hu X, Zeng Q, Xiao J, Qin S, Wang Y, Shan T et al. Herpes simplex virus 1 induces microglia gasdermin D-Dependent pyroptosis through activating the NLR family pyrin domain containing 3 inflammasome. Front Microbiol. 2022;13:838808.

  178. Rogiers O, Frising UC, Kucharíková S, Jabra-Rizk MA, van Loo G, Van Dijck P et al. Candidalysin crucially contributes to Nlrp3 inflammasome activation by Candida albicans Hyphae. mBio. 2019;10:e02221–18.

  179. Goepp M, Crittenden S, Zhou Y, Rossi AG, Narumiya S, Yao C. Prostaglandin E(2) directly inhibits the conversion of inducible regulatory T cells through EP2 and EP4 receptors via antagonizing TGF-β signalling. Immunology. 2021;164:777–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Martínez-Colón GJ, Taylor QM, Wilke CA, Podsiad AB, Moore BB. Elevated prostaglandin E2 post–bone marrow transplant mediates interleukin-1β-related lung injury. Mucosal Immunol. 2018;11:319–32.

    Article  PubMed  Google Scholar 

  181. Ta A, Vanaja SK. Inflammasome activation and evasion by bacterial pathogens. Curr Opin Immunol. 2021;68:125–33.

    Article  CAS  PubMed  Google Scholar 

  182. Ding X, Kambara H, Guo R, Kanneganti A, Acosta-Zaldívar M, Li J et al. Inflammasome-mediated GSDMD activation facilitates escape of Candida albicans from macrophages. Nat Commun. 2021;12:6699.

  183. Elbahlawan L, McArthur J, Quasney MW, Pei D, Srivastava K, Dahmer MK, et al. Association of IL-1β -511 polymorphism with severe veno-occlusive disease in pediatric-matched allogeneic hematopoietic stem cell transplantation. J Pediatr Hematol Oncol. 2012;34:175–9.

    Article  CAS  PubMed  Google Scholar 

  184. Qiao J, Huang Y, Xia Y, Chu P, Yao H, Xu L et al. Busulfan and cyclosphamide induce liver inflammation through NLRP3 activation in mice after hematopoietic stem cell transplantation. Sci Rep. 2015;5:17828.

  185. Inoue Y, Matsunawa M, Sano F, Miura I. Efficacy of recombinant human soluble thrombomodulin in treating disseminated intravascular coagulation complicating allogeneic hematopoietic stem cell transplantation. Acta Haematol. 2018;140:121–7.

    Article  CAS  PubMed  Google Scholar 

  186. Grant A, Chapman LRM, Mitchell R, O’Brien TA. Engraftment syndrome following hematopoietic stem cell transplant: a review of the literature. Clin Transpl. 2020;34:e13875.

    Article  Google Scholar 

  187. Khandelwal P, Mellor-Heineke S, Rehman N, Lane A, Smiley K, Villanueva J, et al. Cytokine Profile of Engraftment Syndrome in Pediatric hematopoietic stem cell transplant recipients. Biol Blood Marrow Transplant. 2016;22:690–7.

    Article  CAS  PubMed  Google Scholar 

  188. Wu B, Jiang C, Jin L, Azadan X, Lin J, Lin L, et al. Serum cytokine profiles during engraftment syndrome and acute graft-versus-host disease in adult patients after hematopoietic stem cell transplantation. Cytokine. 2024;178:156582.

    Article  CAS  PubMed  Google Scholar 

  189. Vande Walle L, Lamkanfi M. Drugging the NLRP3 inflammasome: from signalling mechanisms to therapeutic targets. Nat Rev Drug Discovery. 2023;23:43–66.

    Article  PubMed  Google Scholar 

  190. Zhao Y, Huang J, Li T, Zhang S, Wen C, Wang L. Berberine ameliorates aGVHD by gut microbiota remodelling, TLR4 signalling suppression and colonic barrier repairment for NLRP3 inflammasome inhibition. J Cell Mol Med. 2022;26:1060–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Di Virgilio F, Schmalzing G, Markwardt F. The Elusive P2X7 Macropore. Trends Cell Biol. 2018;28:392–404.

    Article  PubMed  Google Scholar 

  192. Zhong X, Zhu F, Qiao J, Zhao K, Zhu S, Zeng L, et al. The impact of P2X7 receptor antagonist, brilliant blue G on graft-versus-host disease in mice after allogeneic hematopoietic stem cell transplantation. Cell Immunol. 2016;310:71–7.

    Article  CAS  PubMed  Google Scholar 

  193. Pasca S, Jurj A, Petrushev B, Tomuleasa C, Matei D. MicroRNA-155 implication in M1 polarization and the Impact in Inflammatory diseases. Front Immunol. 2020;11:625.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Chen W, Su G, Xu Y, Guo W, Bhansali R, Pan B et al. Caspase-1 inhibition ameliorates murine acute graft versus host disease by modulating the Th1/Th17/Treg balance. Int Immunopharmacol. 2021;94:107503.

  195. Hüber CM, Doisne JM, Colucci F. IL-12/15/18-preactivated NK cells suppress GvHD in a mouse model of mismatched hematopoietic cell transplantation. Eur J Immunol. 2015;45:1727–35.

    Article  PubMed  PubMed Central  Google Scholar 

  196. Song Y, Hu B, Liu Y, Jin Z, Zhang Y, Lin D, et al. IL-12/IL-18-preactivated donor NK cells enhance GVL effects and mitigate GvHD after allogeneic hematopoietic stem cell transplantation. Eur J Immunol. 2018;48:670–82.

    Article  CAS  PubMed  Google Scholar 

  197. Antin JH, Weisdorf D, Neuberg D, Nicklow R, Clouthier S, Lee SJ, et al. Interleukin-1 blockade does not prevent acute graft-versus-host disease: results of a randomized, double-blind, placebo-controlled trial of interleukin-1 receptor antagonist in allogeneic bone marrow transplantation. Blood. 2002;100:3479–82.

    Article  CAS  PubMed  Google Scholar 

  198. Im K-I, Kim N, Lim J-Y, Nam Y-S, Lee E-S, Kim E-J, et al. The free radical scavenger NecroX-7 attenuates Acute Graft-versus-host Disease via Reciprocal Regulation of Th1/Regulatory T Cells and inhibition of HMGB1 release. J Immunol. 2015;194:5223–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  199. Matsuoka S, Hashimoto D, Kadowaki M, Ohigashi H, Hayase E, Yokoyama E, et al. Myeloid differentiation factor 88 signaling in donor T cells accelerates graft-versus-host disease. Haematologica. 2020;105:226–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Hoggatt J, Singh P, Tate TA, Chou B-K, Datari SR, Fukuda S, et al. Rapid mobilization reveals a highly engraftable hematopoietic stem cell. Cell. 2018;172:191–e20410.

    Article  CAS  PubMed  Google Scholar 

  201. Adamiak M, Bujko K, Cymer M, Plonka M, Glaser T, Kucia M, et al. Novel evidence that extracellular nucleotides and purinergic signaling induce innate immunity-mediated mobilization of hematopoietic stem/progenitor cells. Leukemia. 2018;32:1920–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Wilhelm K, Ganesan J, Müller T, Dürr C, Grimm M, Beilhack A, et al. Graft-versus-host disease is enhanced by extracellular ATP activating P2X7R. Nat Med. 2010;16:1434–8.

    Article  CAS  PubMed  Google Scholar 

  203. Ratajczak MZ, Kucia M. Extracellular Adenosine Triphosphate (eATP) and its Metabolite, Extracellular Adenosine (eAdo), as Opposing Yin-Yang regulators of Nlrp3 inflammasome in the trafficking of hematopoietic Stem/Progenitor cells. Front Immunol. 2020;11:603942.

    Article  CAS  PubMed  Google Scholar 

  204. Abdelbaset-Ismail A, Borkowska-Rzeszotek S, Kubis E, Bujko K, Brzeźniakiewicz-Janus K, Bolkun L, et al. Activation of the complement cascade enhances motility of leukemic cells by downregulating expression of HO-1. Leukemia. 2017;31:446–58.

    Article  CAS  PubMed  Google Scholar 

  205. Sauerer T, Velázquez GF, Schmid C. Relapse of acute myeloid leukemia after allogeneic stem cell transplantation: immune escape mechanisms and current implications for therapy. Mol Cancer. 2023;22:180.

  206. Notarantonio A-B, Bertrand A, Piucco R, Fievet G, Sartelet H, Boulangé L et al. Highly immunosuppressive myeloid cells correlate with early relapse after allogeneic stem cell transplantation. Experimental Hematol Oncol. 2024;13:50.

  207. Yang Y, Liu Q, Shi X, Zheng Q, Chen L, Sun Y. Advances in plant-derived natural products for antitumor immunotherapy. Arch Pharm Res. 2021;44:987–1011.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was funded by the National Key Research and Development Program of China (2023YFC2508902), National Natural Science Foundation of China (82070192), Haihe Laboratory of Cell Ecosystem Innovation Fund (22HHXBSS00034), Clinical research project of Tianjin Society of Hematology and Regenerative Medicine (No. 2022 TSHRM08004), and Tianjin Natural Science Foundation (23JCZXJC00220).

Author information

Authors and Affiliations

Authors

Contributions

J.L., M.W. and E.J. conceived the structure of the manuscript . J.L. and Y.Z. drafted the manuscript and prepared the figures. J.Z. prepared the table and assisted in the manuscript preparation. J.L., M.W. and E.J. revised the manuscript. J.L., Y.Z., M.W., J.Z. and E.J. discussed and contributed to the writing of this review. All authors have read and approved the final manuscript.

Corresponding author

Correspondence to Erlie Jiang.

Ethics declarations

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Luo, J., Zhou, Y., Wang, M. et al. Inflammasomes: potential therapeutic targets in hematopoietic stem cell transplantation. Cell Commun Signal 22, 596 (2024). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-024-01974-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12964-024-01974-3

Keywords