Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal inflammatory disease that involves host genetics, the microbiome, and inflammatory responses. The current consensus is that the disruption of the intestinal mucosal barrier is the core pathogenesis of IBD, including intestinal microbial factors, abnormal immune responses, and impaired intestinal mucosal barrier. Cumulative data show that nucleotide-binding and oligomerization domain (NOD)-like receptors (NLRs) are dominant mediators in maintaining the homeostasis of the intestinal mucosal barrier, which play critical roles in sensing the commensal microbiota, maintaining homeostasis, and regulating intestinal inflammation. Blocking NLRs inflammasome activation by botanicals may be a promising way to prevent IBD progression. In this review, we systematically introduce the multiple roles of NLRs in regulating intestinal mucosal barrier homeostasis and focus on summarizing the activities and potential mechanisms of natural products against IBD. Aiming to propose new directions on the pathogenesis and precise treatment of IBD
Keywords: IBD, intestinal mucosal barrier, natural products, NLRs, pathogenesis
Introduction
Inflammatory bowel disease (IBD) is an illness with an unknown etiology and pathogenesis characterized by chronic inflammation of the intestinal mucosa with recurrent and persistently delayed episodes, encompassing ulcerative colitis (UC) and Crohn’s disease (CD). IBD patients are mainly dispersed in developed countries (>0.3%), but the incidence of IBD in newly industrialized countries is rapidly growing (the annual growth rate percentage has reached 14.9%). 1 IBD has thus evolved into a global disease, and the number of IBD patients is expected to reach 30 million by the year 2025. 2 The number of IBD patients is increasing year by year, and there is an urgent need to explore the pathogenesis of IBD and find potential targets and drugs for the treatment of IBD.
IBD is known to be caused by interactions between genetics, host immunity, the mucosal barrier, and the gut microbiome, but its exact etiology remains unclear. 3 Impaired mucosal barrier function is considered to be the primary pathogenic factor in the development of IBD.4,5 As the primary defense barrier of the body, the intestinal mucosal barrier has made outstanding contributions to regulating and maintaining the homeostasis of the internal environment. 6 Once the intestinal mucosal barrier function is disrupted by various factors, the immune balance is disrupted, triggering an inflammatory response that is ultimately involved in the development of IBD. 7 It is believed that NOD-like receptors (NLRs) are the main mediators in maintaining intestinal mucosal barrier homeostasis,8,9 including regulation of intestinal microbiota, inflammatory response, cellular senescence, and cell death. 10 In particular, the role of NLRs in modulating the intestinal mucosal barrier and immune mechanisms has become a hot topic of research and is considered a valuable therapeutic target for the treatment of IBD.
It is thought that aberrant activation of NLRs is the main cause of intestinal mucosal barrier dysfunction. Blocking NLR-inflammasome activation by botanicals or synthetic small molecules may be a valuable way to prevent IBD progression. Natural bioactive compounds are an important resource for drug discovery and modification. Several studies suggest that natural products (NPs), including dietary, and herbal formulations or their extracts, have unique efficacy in the health management of chronic inflammatory diseases.11,12 In this review, we elucidate the composition and function of the intestinal mucosal barrier and discuss the potential role of NLRs in regulating the intestinal mucosal barrier and inflammatory mechanisms. We also summarize the original extracts and traditional medicines targeting NLRs against IBD with the potential to develop NPs.
Literature search strategy
Studies were identified by searching electronic medical databases: Web of Science, PubMed, SciFinder, OvidSP, Scopus, MEDLINE, ScienceDirect, IMBIOMED, and Cochrane database, from January 1983 to February 2023. All human and animal studies are included to evaluate the role of NOD-like receptors in the pathogenesis of IBD, ensuring attention to the latest and comprehensive literature. MeSH terms and/or text words included the broad terms NOD-like Receptors, inflammatory bowel disease, intestinal mucus barrier, and Botanicals. We also searched specific pathogens including NLRP1, NOD2, NLRP3, NLRP6, NLRC4, NOD1, NLRP12, biological barrier, gut microbiota, chemical barrier, mechanical barrier, immunological barrier, botanicals, natural products, flavonoids, terpenoids, plant extracts, alkaloids, phenylpropanoids, treatment, therapeutic, pathogenesis, epidemiology, combined with ‘AND inflammatory bowel disease OR UC OR Crohn disease OR colitis’. We also reviewed the references of relevant IBD disease review articles and studies that met the inclusion criteria of other relevant articles.
The composition and function of the intestinal mucosal barrier
Biological barrier
The gut microbiome is mainly composed of bacteria, archaea, fungi, viruses, and bacteriophages. 13 The gut microbiota is dominated by bacteria, especially members of the phylum Bacteroidetes, Firmicutes, Actinobacteria, and Proteobacteria, the sum of which exceeds 90%. 14 The gut microbiota plays an important role in several aspects of host homeostasis. The gut microbiota plays an important role in several aspects of host homeostasis: (1) Aid the host digest and metabolize indigestible nutrients such as xylan, cellulose, and resistant starch. (2) Provide substrates, enzymes, and energy for host metabolic processes. (3) Prevent the invasion of harmful bacteria by secreting bacterial toxins, producing short-chain fatty acids, promoting intestinal peristalsis, occupying the adhesion sites of pathogenic bacteria, and competing for nutrients. (4) Suppresses the gut immune system’s hyperreactive immune response to antigens to maintain immune tolerance and homeostasis.
Chemical barrier
The intestinal chemical barrier consists of mucus secreted by intestinal epithelial cells, digestive juice fluids (gastric acid, bile), and antimicrobial peptides (AMPs), which cooperate to form a complex protective mechanism. 15 The role of mucus is to insulate intestinal bacteria from contact with the intestinal epithelium, thereby avoiding tissue damage and microbiota translocation. 16 Stomach acid and bile acid could inhibit the growth and reproduction of bacteria, thus protecting the intestinal tract from pathogenic bacteria infection. 17 AMPs can have multiple effects on intestinal mucosal homeostasis; in addition to killing ingested pathogens, controlling the composition of the gut microbiota can also coordinate adaptive immune responses. 18 The intestinal chemical barrier is a good support for the intestinal microbial barrier and plays an important role in maintaining the intestinal microecological balance.
Mechanical barrier
The intestinal mucosal mechanical barrier is composed of intestinal mucosal epithelial cells, the apical junction complex (AJC), and the lamina propria. The AJC is located at the apical junction between adjacent intestinal epithelial cells and is composed of adherens junctions (AJs) and tight junctions (TJs). 19 AJs and TJs include two cell-cell adhesion modes with different functions. The adhesion junction contains intracellular components such as α-catenin, β-catenin, and p120-catenin, as well as the transmembrane protein E-cadherin, which triggers and regulates contact between cells in addition to regulating maturation. 20 TJs are composed of claudin, the transmembrane proteins occludin, and the cytoplasmic scaffolding proteins ZO-1, -2, and -3, which, together, regulate the paracellular pathways of ion and solute movement between cells. 21 Mechanical barriers have several critical physiological functions: (1) Preventing harmful entities in the lumen from invading the body, including foreign antigens, microorganisms, and toxins. (2) Selectively allowing essential dietary nutrients, electrolytes, and water to enter the circulation through the intestinal cavity. (3) Secretion of mucus, antimicrobial proteins (AMPs), and immunoglobulin A (SIgA), which provides the basis for the intestinal chemical barrier, immune barrier, and other barrier functions.
Immunological barrier
The intestinal mucosal immune barrier is mainly composed of gut-associated lymphocyte tissue (GALT) and diffuse immune cells [innate immune cells (neutrophils, monocyte/macrophages, dendritic cells, mast cells, and innate lymphoid cells) and adaptive immune cells (B and T cells)]. 22 Moreover, epithelial cells are increasingly viewed as an integral part of the immune barrier. 23 The GALT secretes immunoglobulin A (IgA) antibodies, and IgA forms IgA – antigen complexes with antigenic substances. The IgA antigen complexes bind to the receptors on M cells. The antigens are then transferred to the lamina propria and subsequently presented to dendritic cells. 24 Antigen-presenting cells in the lamina propria receive immune-stimulating antigens from the cavity, process those antigens, and present them to T cells. The IL-12 secreted by antigen-presenting cells can promote the production of Th1 by T cells, but the simultaneous production of IL-10 limits Th1 immune responses. 25 Additionally, IgA promotes the renewal of intestinal epithelial cells, and repairs damage over time, stabilizing the intestinal mucosal barrier. 26 In summary, the main function of the intestinal mucosal immune barrier is to recognize antigens in the body, generate humoral and cellular immunity, and effectively remove antigens.
In conclusion, the intestinal mucosal barrier is an important protective mechanism and defense system for maintaining body health. Moreover, it is worth mentioning that the interaction between each barrier forms a complex regulatory network, which fully plays the role of preventing the invasion of pathogenic microorganisms, regulating antigen import, and inhibiting inflammation (Figure 1).
Figure 1.
Components of the intestinal mucosal barrier.
NLRs family and signaling mechanisms in the intestinal epithelial barrier
NLRs are composed of multiple domains, including a C-terminal domain, a series of leucine-rich repeats (LRRs), a central nucleotide-binding NACHT domain, and a variable N-terminal effector domain. Specifically, NLRs are divided into five subfamilies based on their unique N-terminal effecter domains: NLRAs with unique acidic activation domains, NLRBs feature a baculovirus inhibitor of apoptosis repeat (BIR)-like domain, NLRCs that have a caspase activation and Death domain or a recruitment domain (CARD), and the NOD and leucine-rich-repeat-containing protein (NLRP) subfamily NLRAs embody a PYRIN domain. 27 The NOD and leucine-rich-repeat-containing protein X (NLRX) subfamily contains only one member, which has an undefined N-terminal domain and has no significant homology with the N-terminal domain of other NLR subfamily members. The LRR domains are responsible for recognizing molecular patterns, including the corresponding components of PAMPs/DAMPs. The characteristic NACHT domain has dNTPase activity and mainly regulates ATP-dependent oligomerization. The N-terminal effector domain is the most prominent component of the NLRs and binds to various linker molecules and downstream effectors to mediate signal transduction.
NLRs are widely expressed in the gut, and their role in the pathogenesis of IBD was previously demonstrated (Table 1).28,29 NOD1 and NOD2 are involved in the recognition of infectious bacteria, induction of immune responses, and protection of the intestinal mucosal barrier from bacterial erosion through the activation of nuclear factor kappa B (NF-κB).30,31 The NLRP3 and NLRC4 assemblages respond to some microorganisms or parasites by activating caspase-1, producing IL-1β and IL-18, and initiating inflammatory processes. NLRP6 recognizes Listeria monocytogenes and recruits caspases, and Gasdermin D (GSDMD) cleavage further enhances NLR’s role as an important molecule in cell death. 32 Similarly, NLRP12 was demonstrated to be crucial in the regulation of interferon and cytokine production, as well as the maintenance of immune homeostasis. 33 Moreover, cell death mediated by NLR-inflammasome activation protects host cells from attack by pathogens, by impeding the nutrient supply to the pathogens, 34 and the activated bystander cells can also secrete antimicrobial agents that arrest pathogen expansion and disease aggravation. 35 However, the specific molecular basis of the causal relationship between NLRs and IBD remains enigmatic. Nonetheless, due to the central roles of NLRs in immune regulatory signaling, they are considered as potential therapeutic targets for IBD.
Table 1.
NLRs in IBD.
| Subfamily | NLR | Cells | Effect | References | |
|---|---|---|---|---|---|
| NLRC | NOD1 |
|
Intestinal epithelial cells and dendritic cells | Protection | Chen et al. 36 Le Bourhis et al. 37 |
| NOD2 |
|
Intraepithelial lymphocyte cells (IELs), enterocytes, Paneth cells, and goblet cells | Protection | Al Nabhani et al. 38 Jiang et al. 39 Richardson et al. 40 Shanahan et al. 41 | |
| NOD2 |
|
/ | Aggravation | Corridoni et al. 42 | |
| NLRC4 |
|
Intestinal epithelial cells and phagocytes | Protection | Sellin et al. 43 Franchi et al. 44 | |
| NLRC4 |
|
/ | Aggravation | Steiner et al. 45 | |
| NLRP | NLRP1 |
|
Intestinal epithelial cells | Protection | Williams et al. 46 |
| NLRP1 |
|
Macrophages | Aggravation | Wang et al. 47 | |
| NLRP3 |
|
Macrophages and neutrophils | Aggravation | Ranson et al. 48 Chen et al. 49 | |
| NLRP3 |
|
Macrophages and neutrophils | Protection | Yao et al. 50 | |
| NLRP6 |
|
Intestinal epithelial cells | Protection | Kempster et al. 51 Yin et al. 52 | |
| NLRP12 |
|
Macrophages and dendritic cells | Protection | Chen et al. 53 | |
![]() LRR PYD NACHT FIIND CARD | |||||
IBD, inflammatory bowel disease.
NOD-like receptors regulate homeostatic intestinal epithelial barrier function
Role of NLRs in modulating intestinal biologic barriers
The gut microbiota is equivalent to a ‘hidden organ’ that helps maintain a healthy mucosal barrier. The composition and diversity of the intestinal microbiota are influenced by host genes.54,55 To assess the contribution of NLRs to the intestinal microbiota in IBD, Elinav et al. analyzed the fecal microbiota based on 16S rRNA. The results showed that the frequency of Prevotella in NLRP6−/− mice were higher. Compared to wild-type mice, NLRP6-/- mice exhibit a higher susceptibility to colitis. 56 Similarly, the study of Seregin et al. demonstrated that NLRP6 could alter Akkermansia muciniphila colonization, preventing IBD, in IL10−/− mice. 57 Interestingly, NLRP6−/− mice are resistant to systemic Escherichia coli, Salmonella typhimurium, and Listeria monocytogenes infections. 58 Gut mucosal microbiome dysbiosis was also reported in mice deficient in NLRP3. Hirota et al. reported that NLRP3−/− mice exhibited enhanced susceptibility to colitis and lower levels of B-defensins compared to wild-type mice, most likely due to altered gut microbiota compositions. Notably, the levels of potentially pathogenic bacteria increased; for example, Enterobacteriaceae, Genus mycobacterium, Collinsella, Subdoligranulum, Clostridium, and Ralstonia increased significantly. 59 However, recent studies contradicted these observations by showing that NLRP3−/− mice were resistant to colitis. 60 Further research showed that mice lacking the NLR components NLRP3, NLRC4, and caspase-1 were highly sensitive to Citrobacter. rodentium-induced gastrointestinal inflammation and could not control C. rodentium due to defects in the generation of IL-18 and IL-1β. 61 Moreover, NLRP3-mutated mice showed reshaping of the gut microbiota and the appearance of regional regulatory T cells (Tregs) to maintain homeostasis and compensate for otherwise-harmful intestinal inflammation. 44 The NLRP3 inflammasome plays a significant role in the pathogenesis of colitis, although NLRP3 was found to have both protective and disadvantageous functions in studies on mucosal immunity. Different results are associated, for example, with the mouse/human genetic backgrounds, 62 according to the composition of the gut microbiota, 63 with different colitis models, 64 or according to the method of inducing colitis (the concentration of DSS, the continuance of DSS delivery, and the number of cycles).
In addition, the regulation of the microbiota of the ileum and the bactericidal activity of ileal crypts are largely determined by NOD2, mainly by altering the interaction between mucosal immunity and ileal microbiota. Biswas et al. reported that NOD2−/− mice presented increased loads of commensal bacteria and pathogens at the terminal ileum. Further analysis showed that NOD2−/− mice experienced a remarkable increase in the amount of Bacteroides, Firmicutes, and Bacillus in the terminal ileum compared to wild-type littermate controls. 65 Notably, increased intestinal inflammation in NOD2-deficient mice and patients with CD was attributed to the impaired bactericidal activity of Paneth cells. 66 Furthermore, reciprocal microbiota transplantation was found to reduce the risk of colitis or colorectal cancer in Nod2−/− mice. 67 Lastly, NLRP12, a negative regulator of inflammation, was identified in an experimental colitis model and patients with IBD. 68 NLRP12 suppresses the excessive production of inflammatory cytokines in the intestines and decreases the susceptibility to colitis by maintaining commensal variety and protective microbiota. The sequencing of bacterial 16S rRNA confirmed that NLRP12−/− may result in the loss of protective gut commensal strains (Lachnospiraceae) and an increase in Escherichia coli strains (Erysipelas), which can be reversed via the administration of beneficial commensal Lachnospira isolates or antibodies targeting inflammatory cytokines. Additionally, NLRP12-deficient mice could be treated with microbiome transplants or antibodies targeting inflammatory cytokines. 53 The above studies on the microbiome in NLRP12-deficient mice resemble those of human IBD patients.69 -71
To date, accumulating evidence has demonstrated the impact of NLRs on the gut microbiota and IBD and illustrated the potential of manipulating the gut microbiota in the treatment of IBD. Therefore, there is great interest in the possible benefits of microbiome-modulating interventions, such as probiotics, prebiotics, antibiotics, fecal microbiota transplantation, and the role of genetic manipulation in the treatment of IBD. However, the underlying mechanisms of how genetic alterations affecting NLRs affect gut microbiota biodiversity are still not fully understood. Additionally, the relationship between gut dysbiosis and IBD is complex and dynamic, rather than a simple causal relationship.
Role of NLRs in modulating the intestinal mechanical barrier
Gut mechanical barrier dysfunction is a consistent feature of IBD. 72 This barrier is composed of intact intestinal epithelial cells, the AJC, and the lamina propria. 73 AJC maintains intestinal homeostasis mainly by controlling the permeability of intestinal epithelial cells to limit the transport of relatively large molecules.74,75 Accumulating data support a correlation between NLRs and the integrity of the intestinal mechanical barrier. protects mice against increased intestinal mucosal permeability caused by AJC dysfunction by regulating myosin light chain kinase (MLCK) signaling76,77 In Caco-2, a small intestinal eNOD2pithelial cell similar in structure and function to differentiation, NLRP3 weakens the cell’s trans epithelial resistance by reducing the expression of claudin-1, occludin, and ZO-1, while increasing the expression of claudin-2 and repositioning occludin and ZO in Caco-2 monolayer. 78 Likewise, the NLRP3 inflammasome was activated in antibiotic-exposed Caco-2 and mice, resulting in decreased expression of claudin and disrupted ZO-1 morphology. 79 In addition to the destruction of AJC, abnormal death of intestinal epithelial cells is a major cause of intestinal mucosal mechanical barrier dysfunction. 80 Autophagy has been shown to regulate cytokine-induced programmed cell death in intestinal epithelial cells, limiting intestinal inflammation. The inflammasome and autophagy are mutually regulated, and this bidirectional regulation provides the necessary checks and balances between host defenses against inflammatory responses and the prevention of excessive inflammation in organ and tissue damage and inflammatory diseases. NLRP6 deficiency results in defective autophagy in goblet cells and reduced mucin secretion in the lumen of the large intestine. 81 When NLRP6-deficient mice were treated with DSS, they became more susceptible to serious colitis due to a reduction in IL-18 expression by intestinal epithelial cells (IECs), and pathological damage increased compared to wild-type animals. 82 Akin to NLRP6, NLRP3 performs a protective function for intestinal epithelial cells. Compared to wild-type mice, NLRP3-deficient mice are more likely to suffer from C. rodentium infections. 83 Simultaneously, activating the NLRP3 inflammasome can induce the compensatory proliferation of epithelial cells, protecting the integrity of the epithelial layer during DSS-induced colitis. 84 NLRC4 is a double-edged sword. On the one hand, NLRC4 can restrict infections by S.typhimurium by activating pyroptosis and forcing infected intestinal epithelial cells to enter the lumen. 43 On the other hand, excessive NLRC4 results in too much pyroptosis, which triggers small intestinal damage. 85
Role of NLR in regulating mucosal immunity
The intestine serves as the largest immune organ in mammals, containing 90% of the immunocompetent cells. The intestinal mucosal immune barrier comprises gut-associated lymphoid tissues (such as Peyer’s patches and mesenteric lymph nodes), cells [such as innate immune cells (neutrophils, monocyte/macrophages, dendritic cells, mast cells, and innate lymphoid cells) and adaptive immune cells (B and T cells)], and molecules (such as immunoglobulins and AMPs). 22 Moreover, epithelial cells are increasingly viewed as an integral part of the immune barrier.
NLRP1 was the first pattern recognition receptor (PRR) discovered to form an inflammasome, and genome-wide association studies have identified NLRP1 mutations linked with IBD. 86 Previous evidence showed that NLRP1b−/− mice showed a significant increase in the incidence of colitis, infections, and colitis-related tumorigenesis via the modulation of IL-1β and IL-18 levels. Moreover, the wound healing and defense responses in DSS-induced colitis and lipopolysaccharide (LPS)-stimulated cellular inflammation in mouse macrophages might be caused by the downregulation of NLRP1 expression. 46 Similarly, gut-barrier defects and excessive intestine-related lymphoid tissue activation are features of NOD2 deficiency in humans and mice. 38 Further investigations revealed that hyperresponsive macrophages and colitis promoted NOD2 signaling in IL-10-deficient mice, and a deficiency of NOD2 ameliorated chronic colitis in IL-10−/− mice. 87 Additionally, NOD2−/− can lead to a steady-state imbalance, inactivated fibroblasts, and inflammatory macrophages, manifested by the increased expression of aberrant activation-related transcriptomes caused by the upregulation of upstream transcription factors such as WT1 and STAT3, and it causes the gp130 family to upregulate the expression of certain genes (IL-11, IL-6, OSM, etc), thereby increasing the risk of intestinal stenosis. 88 Moreover, NLRP3 is the most widely studied immune regulator in NLRs, although its role is controversial. Chen et al. showed that the interaction of the NLRP3 inflammasome with NEK7 promotes IBD by mediating NF-κB signaling to regulate macrophage apoptosis. 49 By contrast, NLRP3-inflammasome activation can facilitate the restoration and reproduction of the intestinal mucosa, which could be related to enhanced levels of caspase-1, IL-1β, and IL-18. 89 IL-1β promotes the production of IL-22 and IL-10 to maintain epithelial integrity.90,91 IL-18 and IL-1β were shown to promote compensatory epithelial cell proliferation and reduce DSS-induced inflammatory colon injury. 83 Intriguingly, another study showed that uncontrollably active NLRP3 inflammasomes neutralize intestinal inflammation by inducing the production of Tregs and reshaping the gut microbiota in NLRP3-mutant mice. 50 In addition to the above NLRs, NLRP6 is also an important regulator of IBD. NLRP6−/− mice showed an increase in the number of CD45+cells in the lamina propria, proliferation of colonic crypts, enlargement of Peyer’s patches, and formation of germinal centers. 56 These mice exhibited increased mucosal permeability and were unable to recover from colitis.82,92
In summary, NLRs play critical roles in immune cell development and function through multiple canonical immune regulatory mechanisms, including caspase-1, RIP2 kinase, mitogen-activated protein kinase (MAPK), and NF-κB signal pathway. However, with the deepening of the research on NLRs in IBD, the functions of these proteins have also become more diverse, but the exact roles of some NLRs in IBD are still controversial and deserve further study. In addition, the biochemical processes regulating NLRs activation and downstream functions remain to be further investigated.
Targeting NLRs with botanicals in IBD therapy
Many synthetic drugs and monoclonal antibodies are currently administered in the clinic to treat IBD, such as NF-κB inhibitors (e.g. sulfasalazine), TNF-α antagonists (e.g., infliximab), and glucocorticoids (e.g. prednisolone). However, the therapeutic effects of these drugs are unsatisfactory, as their long-term administration can produce severe side effects, leading to poor patient acceptance and compliance.93–95 Therefore, exploring safe and effective drugs has become an urgent task for the treatment of IBD patients.
NLRs are important regulators of intestinal mucosal homeostasis and are promising therapeutic targets for IBD. NLRs have regulatory effects on multiple signaling molecules explicitly related to IBD diseases, such as NF-κB, ASC, caspase-1, MAPK, IL-1β, TNF-α, IL-6and IL-18 (Figure 2). The apparent breadth and magnitude of NLRs’ involvement illustrate the apparent centrality of this protein family. As of November 2019, 82 patents were pending for potential therapeutic NLRs modulators, and 22 patents had been granted. 96 Additionally, there are multiple phase I clinical trials of NLRP3 inhibitors for CD that have been registered in public databases by Novartis. 97
Figure 2.
The mechanism of NLRs in the pathogenesis of IBD.
NPs, including dietary compounds, herbal formulas, and their extracts, have been known to have unique curative effects in the management of chronic inflammatory diseases for thousands of years. Numerous experiments in vitro and in vivo have indicated that some active ingredients in botanicals and NPs perform anti-IBD functions by regulating NOD-like receptor signaling, with few toxic side effects compared to synthetic treatments. In this section, we review the current findings on targeting NLRs with botanicals in IBD intervention (Table 2).
Table 2.
Targeting NLRPs with botanicals in IBD therapy.
| Agent | Experimental subjects | Outcomes | Potential mechanisms | References |
|---|---|---|---|---|
| Wogonoside | Female BALB/c mice (DSS), THP-1 cells | Inflammatory cytokines↓, inflammatory cell infiltration↓, colonic pathological damage↓ | Blocking activation of NF-κB and NLRP3 inflammasome. | Sun et al. 98 |
| Flavonoid VI-16 | Male C57BL/6 J mice (DSS), Macrophages | Colonic pathological damage↓, inflammatory cytokines↓ | Suppressing ROS/Txnip/NLRP3 signaling pathway. | Zhao et al. 99 |
| Curcumin | Female C57BL/6 mice (DSS), Macrophages | DAI↓, inflammatory cytokines↓, colonic pathological damage↓ | Blocking activation of NLRP3 inflammasome and ASC oligomerization. | Gong et al. 100 |
| Procyanidin | Male C57BL/6 mice (DSS), Macrophages | Colonic pathological damage↓, inflammatory condition↓ | Suppressing activation of MMP9, NF-κB, and NLRP3 inflammasome. | Chen et al. 101 |
| Naringin | Male C57BL/6 mice (DSS), RAW264.7 cells | Inflammatory cytokines↓, disease severity↓, DAI↓ | Activating PPARγ and suppressing NF-κB, MAPK, and NLRP3-inflammasome activation. | Cao et al. 102 |
| Phloretin | Male C57BL/6 mice (DSS) | Escherichia coli and Lactobacillus levels rebalanced, gut epithelial barrier integrity↑ | Upregulating ZO-1 and occludin expression, inhibiting NLRP3 inflammasome activation. | Zhang Z et al. 103 |
| Genistein | C57BL6/J mice (DSS), Macrophages | Pro-inflammatory mediator production↓, disease severity↓, inflammatory cell infiltration↓ | Inhibiting NLRP3 inflammasome by regulating TGR5-cAMP pathway. | Chen et al. 104 |
| Cardamonin | Male C57BL/6 mice (DSS), Male BALB/c mice (TNBS:2,4,6-trinitrobenzenesulfonic acid) |
Myeloperoxidase activity↑, DAI↓, colonic pathological damage↓ | Activating AhR/Nrf2/NQO1 pathway and inhibiting NLRP3-inflammasome activation. | Wang et al. 105 |
| Alpinetin | Female BALB/c mice (DSS), THP-1 cells |
Diarrhea and colonic histological injury↓, inflammatory cytokines↓ | Downregulating TLR4, NF-κB, and NLRP3-inflammasome expression. | He et al. 106 |
| Formononetin | Male C57BL/6 mice (DSS), HCT-116 cells |
Epithelial cell injury↓, disease severity↓ | Increasing tight junction proteins expressions and inhibiting the NLRP3 pathway. | Liu et al. 107 |
| Apigenin | Male C57BL/6 mice (DSS) | Inflammatory cytokines↓, colonic pathology damage↓. | Reprograming the gut microbiota through regulating NLRP6. | Radulovic et al. 108 |
| Paeoniflorin | C57BL/6 mice (DSS), RAW264.7 cells | Gram-positive bacteria count and gram-positive bacterial infiltration↓, disease severity↓. | Inhibiting gram-positive bacteria-dependent MDP–NOD2 pathway. | Luo et al. 109 |
| Toosendanin | Male C57BL/6 mice (DSS) | M1 macrophage polarization↓, disease severity↓ | Upregulating HO-1/Nrf2 expression and inhibiting NLRP3 inflammasome activation. | Fan et al. 110 |
| Asiatic acid | Female C57BL/6 mice (DSS), THP-1 cells | Inflammatory cytokines↓, disease severity↓ | Suppressing mitochondria-mediated NLRP3-inflammasome activation. | Guo et al. 111 |
| Dihydroartemisinin | Male C57BL/6 mice (DSS) | DAI↓, inflammatory cytokines↓, disease severity↓ | Inhibiting the phosphorylation of NF-κB p65 and p38 MAPK, and NLRP3-inflammasome activation. | Liang et al. 112 |
| Ginsenoside Rg1 | C57BL/6 mice (DSS) | DAI↓, pro-inflammatory cytokines levels↓, colonic damage scores↓. | Activating TLR4-NLRP12-NF-κB pathway. | Zhu et al. 113 |
| Celastrol | Male SD rats (DSS) | DAI and MDI↓, colonic histological damage↓, inflammatory cytokines↓ | Modulating HSP90 and inducing autophagy to enhance NLRP3 sensitivity to CP-456773. | Saber et al. 114 |
| Libertellenone M | Female C57/BL6 mice (DSS), RAW264.7 cells. |
DAI↓, disease severity↓, inflammatory cytokines↓. |
Inhibiting the nuclear translocation of NF-κB and the assembly of NLRP3 inflammasome | Fan et al. 115 |
| Ginsenoside Rk3 | male C57BL/6 mice (DSS) | Clinical symptoms↓, inflammatory cell infiltration↓, pro-inflammatory cytokines↓, | Inhibiting NLRP3-inflammasome activation | Tian et al. 116 |
| Palmatine | Male BALB/C mice (DSS), THP-1 cells | DAI↓, colonic inflammation↓, disease severity↓. | Facilitating PINK1/Parkin-driven mitophagy-mediated NLRP3-inflammasome inactivation. | Mai et al. 117 |
| Fumigaclavine C | female C57BL/6 mice (DSS), THP-1 cells | Disease severity↓, inflammatory cytokines↓, DAI↓. | Suppressing NF-kB, STAT3, STAT1, and NLRP3-inflammasome activation. | Guo et al. 118 |
| Evodiamine | Male C57BL/6 mice (DSS) | DAI↓, Escherichia coli, and Lactobacillus levels rebalanced, colonic pathological damage↓. | Suppressing NF-κB and NLRP3-inflammasome activation. | Shen et al. 119 |
| Carboxyamidotriazole | Female SD rats (TNBS) | Intestinal inflammatory and permeability↓, colonic pathological damage↓. | Inhibiting NLRP3-inflammasome and NF-κB activation. | Du et al. 120 |
| Compound 1 | Female C57BL/6 mice (DSS), THP-1 cells | Mucosal damage↓, inflammatory cell infiltration↓, CD11b+ infiltrating cells↓ | A novel potent Nrf2/ARE inducer, inhibiting NLRP3-inflammasome activation. | Wang et al. 121 |
| Chlorogenic acid | BALB/c mice (DSS) RAW264.7 cells. |
Colonic inflammation↓, disease severity↓. | Suppressing miR-155/NF-κB/NLRP3 pathway. | Zeng et al. 122 |
| Bergenin | Male Wistar rats (TNBS) | MPO↓, inflammatory cytokines↓, Disease severity↓. | Blocking pSTAT3, NF-κB canonical, and NLRP3/ASC pathways. | Lopes de Oliveira et al. 123 |
| Brusatol | Male SD rats (TNBS), RAW264.7 cells. | Colonic pathology damage↓, oxidative stress↓, DAI↓. | Suppressing NF-κB and NLRP3 activation and enhancing Nrf2 expression | Zhou et al. 124 |
| Fraxinellone | Male C57BL/6 mice (DSS), THP-1 cells | Macrophage infiltration↓, colonic pathology damage↓, inflammatory cytokines↓. | Suppressing NF-κB signaling and NLRP3 inflammasome activation. | Wu et al. 125 |
| VitD3 | Male C57BL/6 mice (DSS), Macrophages | Macrophage infiltration↓, colon inflammation↓, CD4+ T cells generation↓. | Inhibiting VDR/ NLRP3/ASC pathway and NLRP3-NEK7 interaction. | Cao et al. 126 |
| MLB | Male C57BL/6 J mice (DSS). | Colonic pathology damage↓, inflammatory cytokines↓. | Inhibiting the activation of NRLP3/ASC/caspase-1 pathway. | Jiang et al. 127 |
| Physalin B | Male BALB/c mice (DSS), RAW 264.7 cells | Colonic pathology damage↓, inflammatory cytokines↓, DAI↓. | Suppressing the activation of STAT3, β-arrestin1, and NLRP3 inflammasome. | Zhang et al. 128 |
| Oryzanol | SD rats (DSS), Caco-2 cells | Gut-barrier damage of colitis rats↓abundances of Alloprevotella, Roseburia, Treponema, Muribaculaceae, and Ruminococcus↑. | SuppressingTLR4/NF-κB/NLRP3 signaling axis. | Xia et al. 129 |
| Munronoid I | Female wild-type C57BL/6 mice, Mouse peritoneal macrophages, MODE-K cells | Colonic pathology damage↓, disease severity↓. | Promoting K48-linked ubiquitination and degradation of NLRP3 | Ma et al. 130 |
| Artemisinin analog SM934 | Female C57BL/6 mice, HT-29 and Caco-2 cells. | Intestinal barrier function↑, intestinal epithelial cell apoptosis↓. | Suppressing NLRP3/NF-κB/MAPK signal axis | Shao et al. 131 |
| Soy isoflavones | Male C57BL/6 mice (DSS), NCM460 cells | Pro-inflammatory cytokines↓, colonic pathology damage↓. | Inhibiting ERα/NLRP3 inflammasome activation. | Gao et al. 132 |
| LEP | Male ICR mice | Oxidative stress damage↓, disease severity↓. | Regulating PPARγ/NF-κB and IL-6/STAT3 pathways and inhibiting NLRP3 inflammasome. | Zong S et al. 133 |
| QCWZD | Male SD rats (DSS) | Butyricimonas, Blautia, and Odoribacter↑, disease severity↓. | Inhibiting the activity of the TLR4/Blimp-1 pathway to promote NLRP12 expression | Sun et al. 134 |
| QRJPD | Male C57BL/6 J mice | Colonic pathology damage↓, disease severity↓. | Inhibit activation of the NLRP3 inflammasome | Zhang et al. 135 |
| APS | Male C57BL/6 mice (DSS) | Colonic pathology damage↓, disease severity↓. | Inhibiting the NLRP3 signaling pathway | Tian et al. 136 |
| LEM | Male C57BL/6 mice (DSS) | DAI↓, Inflammatory cell infiltration↓, intestinal barrier integrity↑. | Modulating NF-κB and MAPK pathways and Inhibiting NLRP3 inflammasome activation. | Zong et al. 137 |
| RGE + EKN | C57BL/6 mice (DSS). | DAI↓, oxidative stress↓, pro-inflammatory cytokines↓, intestinal mucosal function↑. | Inhibiting TLR4/ NF-κB and NLRP3 inflammasome pathways | Saba et al. 138 |
| CGB | C57BL/6 mice (DSS) | Inflammatory cell infiltration↓, colonic pathology damage↓. | Suppressing the NLRP3 signaling pathway and restores the expression of occludin and claudin-1. | Mahmoud et al. 139 |
| Platycodon grandiflorum root | Macrophages, Intestinal epithelial cells, Mice (DSS) | Inflammatory cytokines↓,colonic pathology damage↓. | Inhibiting NF-κB signaling pathway and the expression of NLRP3 inflammasomes by activating AMP-activated protein kinase | Wang et al. 140 |
APS, astragalus polysaccharide; CGB, Canna × generalis L.H. Bailey; EKN, epimedium Koreanum Nakai; LEM, lycium ruthenicum Murray; LEP, lachnum polysaccharide; MAPK, mitogen-activated protein kinase; MLB; Magnesium lithospermate B; NF-κB, nuclear factor kappa B; RGE, Red ginseng extract; QCWZD, Qingchang Wenzhong decoction; QRJPD, Qingrejianpi decoction.
Flavonoids
Flavonoids have been reported to encompass multiple biological activities, including immune adjustment, scavenging free radicals, the changing of the microbiota constitution, antiapoptotic, and the restoring of damaged epithelial barriers. For this reason, flavonoids may be a promising approach for IBD management. For instance, wogonoside, a natural flavonoid isolated from Scutellaria baicalensis, attenuates DSS-induced colitis in mice through the inhibition of NF-κB and NLRP3 inflammasome. 98 Similarly, Flavonoid VI-16 (a synthetic flavonoid compound), selectively inhibits Txnip-dependent activation of NLRP3 inflammasome to alleviate oxidative stress in colonic macrophages, thereby reducing DSS-induced colitis. 99 Other flavonoids NLRP3 signaling inhibitors, such as curcumin, procyanidin, naringin, phloretin, cardamonin, alpinetin, genistein, and formononetin, have likewise been shown to be effective in the treatment of colitis.100 –107 Other flavonoid NLRs inhibitors also have a potentially preventive or therapeutic effect on colitis. Apigenin, a common dietary flavone, protects against gut inflammation by modulating the intestinal microbiota constituents through NLRP6 and C-type lectin Reg3b in mice. 108
Terpenoids
Terpenoids constitute the largest class of NPs and are a rich reservoir of candidate compounds for drug discovery. Several preclinical studies have provided evidence that terpenoids are useful in IBD. Paeoniflorin, a natural monoterpenoid isolated from Paeonia lactiflora Pall, was proven to facilitate intestinal mucosal barrier protection in a DSS-induced colitis model. Detailed studies indicated that paeoniflorin ameliorates DSS-induced colitis in mice by decreasing the infiltration of gram-positive bacteria through the regulation of the gram-positive-bacterium-dependent MDP–NOD2 pathway. 109 Toosendanin is derived from the fruits or bark of Melia toosendan Sieb. Et Zucc (Meliaceae), potently blocked NLRP3 inflammasome activation via Nrf2/HO-1 signaling in mouse IBD models. 110 Asiatic acid was demonstrated to block the ROS–NLRP3–caspase-1–IL-1β cascade in macrophages and experimental mouse colitis models. 111 Dihydroartemisinin prevents DSS-induced colitis in mice by effectively blocking p38 MAPK signaling and NLRP3-inflammasome activation. 112 Ginsenoside Rg1, a pharmacologically active component of ginseng, effectively blocked the expression of NLRP12 and NF-κB signaling and reduced the inflammation of DSS-induced colitis in mice. 113 Celastrol, a natural triterpenoid extracted from the root bark of Tripterygium wilfordii Hook. f. Celastrol, protected mice against colitis by inhibiting the activation of the NLRP3 inflammasome signaling. 114 Libertellenone M, a hippocampal diterpenoid from the marine-derived fungus Stibella fimetaris, inhibited NLRP3-inflammasome activation in a mouse model of IBD. 115 In addition, Ginsenoside Rk3, selectively blocked NLRP3 inflammasome activation, leading to reduced TNF-α, IL1β, and IL-6 production in a mouse model of IBD. 116
Alkaloids
An increasing number of recent studies have reported that alkaloids are effective in treating intestinal inflammatory disorders. Palmatine, originally isolated from medicinal rayon fibers, promotes PINK1/Parkin-mediated mitochondrial autophagy and inhibits NLRP3 inflammatory vesicle activation, leading to a decrease in IL-1β, thereby alleviating DSS-induced colitis in mice. 117 Fumigaclavine C, isolated from Aspergillus fumigatus cultures, inhibits the formation of the NLRP3 inflammasome to alleviate DSS-induced colitis. 118 Evodiamine, an alkaloidal compound isolated from the traditional Chinese medicine Evodia rutaecarpa (Juss.), exerts therapeutic activities in treating DSS-induced UC mice by regulating NF-κB signaling and the NLRP3 inflammasome and inhibiting the production of TNF-α, IL-1β, and IL-6. 119 Carboxyamidotriazole, an inhibitor of non-voltage-gated calcium channels, maintains intestinal integrity by inhibiting the expression of inflammatory cytokines, NF-κB signaling, and NLRP3-inflammasome activation in TNBS-induced mice. 120 3-(2-Oxo-2-phenylethylidene)−2,3,6,7-tetrahydro-1H-pyrazino[2,1-a]isoquinolin-4(1 1bH)-one (compound 1), a new potential Nrf2/ARE inducer, reduces DSS-induced inflammatory cell infiltration and colon pathological damage by activating Nrf2 to block the NLRP3 initiation step. 121
Phenylpropanoids
Phenylpropanoids are attractive molecules for the development of new drugs against a wide range of diseases, such as those related to inflammatory processes. In IBD, compounds from the plant phenylpropanoid metabolic pathway have yielded promising associations. Chlorogenic acid, a dietary polyphenol from the hydroxycinnamic acid family, widely exists in medicinal herbs and human diets. Zeng et al. found that chlorogenic acid can maintain intestinal microecology and immune balance by suppressing NLRP3-inflammasome-related signaling in DSS-induced mice. Furthermore, chlorogenic acid inhibits RAW264.7 cell activation by reducing the expression of p-NK-κB and NLRP3-inflammasome proteins, which depend on the downregulation of miR-155 expression. 122 Bergenin, a natural secondary metabolite abundant in Shiso Bergen, Mallotus japonicus, and Gabonese Shagroti, prevents non-classical and classical NLRP3/ASC inflammatory access by regulating pSTAT3 and NF-κB signaling to reverse TNBS-induced colon tissue damage in acute colitis in mice. 123 Brusatol, a quassinoid compound isolated from Brucea javanica, has protective effects against TNBS-induced colitis that are closely related to the mechanism through inhibition of NF-κB and NLRP3-mediated inflammatory responses and regulation of Nrf2-mediated oxidative stress.124,125
Other compounds
Apart from the NPs mentioned above, many other polysaccharides, proteins, amino acids, and other active NPs also exert anti-IBD biological activities by regulating NLR-mediated inflammatory responses. Fraxinellone, a product of limonoids originating from the root bark of Dictamnus dasycarpus, potently blocked the activation of NLRP3 inflammasome and NF-κB signaling in macrophage activation and alleviate intestinal inflammatory responses. 126 1,25-Dihydroxy Vitamin D3 (VitD3) is a steroid hormone with anti-proliferative/immunomodulatory effects, inhibited the oligomerization of the caspase recruitment domain (ASC) and specifically inhibits the activation of NLRP3. Subsequently, NLRP3 was found that NLRP3 mediates the inhibition of vitamin D receptors on caspase 1 activation and IL-1 secretion in DSS-induced UC mice. 127 Magnesium lithospermate B is a salvianolic compound derived from Salvia miltiorrhizae radix, which protects the colonic mucosa from acute and chronic colitis by inhibiting the NLRP3/ASC/caspase-1 pathway in DSS-induced mice. 128 Physalin B is a withanolide compound abundant in Physalis alkekengi L. var. franchetii (Mast.) Makino (a nutraceutical and Chinese herbal medicine), ameliorates general symptoms and colon pathological damage by inhibiting the activation of STAT3, β-arrestin1, and NLRP3 inflammasomes in DSS-induced colitis mice. 129 Oryzanol, a nutrient component of cereal grains, attenuates intestinal barrier damage and inflammatory responses by modulating the DSS-induced intestinal microbiota-TLR4/NF-κB/NLRP3 signaling axis in UC rats. 130 Monroxone I is a diterpenoid compound isolated and purified from the Meliaceae family that inhibited pro-inflammatory factor production and typical apoptosis of mouse peritoneal macrophages by promoting k48 ubiquitination and NLRP3 degradation in DSS-induced mice. 131 Artemisinin analog SM934, a new water-soluble artemisinin analog, protects mice with colitis from TNBS-induced intestinal barrier disruption by inhibiting epithelial cell apoptosis and focal death through regulation of the NLRP3/NF-κB/MAPK signaling axis. 132
Plant extracts
Except for isolated phytochemicals, other botanicals and extracts may also have potential preventive or therapeutic effects on IBD driven by NLRs. Soy isoflavones (SIFs) are selective estrogen receptor modulators with anti-inflammatory activity. In IBD, SIFs ameliorate DSS-induced colitis by inhibiting ERα expression and the NLRP3-inflammasome pathway. 133 Lachnum polysaccharide (LEP) alleviates DSS-induced colitis by suppressing NLRP3 activation and downregulating ASC, caspase-1, and IL-1β levels. 134 Qingchang Wenzhong Decoction (QCWZD) is a traditional Chinese herbal formula widely used in clinical practice for the treatment of IBD. QCWZD protected against IBD by inhibiting TLR4/Blimp-1 pathway activity promotes NLRP12 expression and selectively ameliorates beneficial strains such as Odoribacterium, Braunschweiger, and Clostridium butyricum while reducing intestinal pathogenic bacteria, including Dora and Clostridium reaching. 135 Qingrejianpi Decoction can reduce NLRP3 expression, reduce the secretion of inflammatory cytokines, and inhibit inflammatory infiltration of immune cells to inhibit the occurrence and development of IBD. 136 Astragalus polysaccharide, the main active ingredient of Astragalus membranaceus, effectively attenuated colitis in mice by inhibiting the expression of apoptotic point proteins containing the C-terminal caspase collecting domain, caspase-1, NOD-like receptor protein 3, IL-18, and IL-1β. 137 Lycium ruthenicum Murray is an edible ethnomedicinal and nutritional food that inhibits the NF-κB and MAPK pathways, suppresses NLRP3 inflammasome activation, and restores intestinal immune homeostasis by enhancing intestinal antioxidant defenses. 138 Red ginseng extract (RGE) +Epimedium Koreanum Nakai (EKN) synergistically reduce the plasma and tissue levels of cytokines and pro-inflammatory mediators by inhibiting the expression of NF-κB of activated B cells and the nucleotide-binding domain of NLRP3 in DSS-induced colitic mice. In vitro, RGE + EKN can dramatically decrease the levels of nitric acid (NO), cytokines, and pro-inflammatory mediators by downregulating the expression of MAPK and NF-κB in RAW 264.7. 139 Canna × generalis L.H. Bailey (CGB) is a traditional anti-inflammatory agent. Studies have confirmed that CGB rhizome extract restores DSS-induced intestinal mucosal dysfunction, reduces oxidative stress, and suppresses inflammatory responses in mice with colitis by inhibiting the NLRP3 inflammasome and TLR4/NF-κB pathways. 140 Platycodon grandiflorus is a dual-use herb for medicinal and food purposes. The fermentation solution of Platycodon grandiflorus root reduces the level of inflammatory factors in serum and the expression of inflammatory factor mRNA in the intestine by inhibiting the AMPK/NF-κB/NLRP3 pathway, regulates the polarization of M1/M2 macrophages, and increases the expression of tight junction protein mRNA in intestinal epithelial cells in a mouse model of IBD. 141
Conclusions and perspectives
IBD has become a serious public health problem affecting human health and is becoming increasingly prevalent around the world. Due to its therapeutic difficulties and the need for long-term treatment, IBD has become a hot topic in the global medical field. The current consensus is that disruption of the intestinal mucosal barrier leading to the displacement of bacteria, which in turn causes a series of immune and inflammatory responses is thought to be the pathogenesis of IBD. Repairing the intestinal mucosal barrier is a highly coordinated process that involves the intestinal flora, TJs, inherent immune recognition, and self-renewal of epithelial cells. Several recent studies highlighted the important benefits of NLRs for gut health.
NLRs maintain the intestinal mucosal barrier’s integrity and homeostasis by controlling the diversity and stability of the gut microbiome, modulating inflammatory signaling events, and amplifying IEC crosstalk with immune cells. Experimental and clinical evidence indicates that NLRs mediate the establishment and homeostatic regulation of the intestinal mucosal barrier. NLRs have become an important target for the treatment of IBD and drug screening. However, diverse mechanisms may contribute to mucosal cures via NLRs. Consequently, there is a demand for further research to specifically illustrate the benefits of the various NLR-mediated signal-conduction mechanisms in the moderation of gut homeostasis and IBD.
This review focused on NPs that can modulate NLRs, making them promising candidates for the development of lead structures in anti-IBD drugs. However, most NPs targeting NLRs lack isoform selectivity, which is important for determining their potential use as therapeutic agents. Nevertheless, the structure types presented in this review offer a well-founded basis indicating that screening and chemical modifications of NPs will, in the future, identify not only more specific inhibitors with fewer adverse side effects but also important characteristics for the elucidation of NLRs’ functions concerning IBD treatment.
In the future, further studies are needed to explain the precise molecular modes of the functions of these botanicals in the activation of NLRs. In addition, the potential toxicities and drug-to-drug interactions of these botanicals in the human body should be explored. Furthermore, the anti-IBD effects of botanicals have not been clinically verified. Generally speaking, since the presence of NLRs is important in both the development and progression of IBD, addressing related NLR modulators is a promising starting point for developing new and potent anti-IBD drugs.
Acknowledgments
None.
Footnotes
ORCID iD: Gang Pei
https://orcid.org/0000-0002-3389-7868
Contributor Information
Feng Zhou, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha, China.
Guo Dong Zhang, Ningxia Medical University, Ningxia, China.
Yang Tan, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Science and Technology Innovation Center/State Key Laboratory Breeding Base of Chinese Medicine Powder and Innovative Medicine, Hunan University of Chinese Medicine, Changsha, China.
Shi An Hu, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China; Hunan Provincial Key Laboratory of TCM Prevention and Treatment of Depression Diseases, Changsha, China.
Qun Tang, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Medical School, Hunan University of Chinese Medicine, Changsha, China.
Gang Pei, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; Key Laboratory of Modern Research of TCM, Education Department of Hunan Province, Changsha, China.
Declarations
Ethics approval and consent to participate: Not applicable.
Consent for publication: Not applicable.
Author contributions: Feng Zhou: Writing – original draft; Writing – review & editing.
Guo Dong Zhang: Investigation; Writing – review & editing.
Yang Tan: Investigation; Resources.
Shi An Hu: Software.
Qun Tang: Data curation; Supervision.
Gang Pei: Funding acquisition; Supervision.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was supported by the National Natural Science Foundation of China, No 82174271.
The authors declare that there is no conflict of interest.
Availability of data and materials: Not applicable.
References
- 1. Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet 2017; 390: 2769–2778. [DOI] [PubMed] [Google Scholar]
- 2. Porter RJ, Kalla R, Ho GT. Ulcerative colitis: recent advances in the understanding of disease pathogenesis. F1000Research 2020; 9: 294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Turpin W, Goethel A, Bedrani L, et al. Determinants of IBD Heritability: genes, bugs, and more. Inflamm Bowel Dis 2018; 24: 1133–1148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Spalinger MR, Sayoc-Becerra A, Ordookhanian C, et al. The JAK inhibitor tofacitinib rescues intestinal barrier defects caused by disrupted epithelial-macrophage interactions. J Crohns Colitis 2021; 15: 471–484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Lei-Leston AC, Murphy AG, Maloy KJ. Epithelial cell inflammasomes in intestinal immunity and inflammation. Front Immunol 2017; 8: 1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Sun X, Yang Q, Rogers CJ, et al. AMPK improves gut epithelial differentiation and barrier function via regulating Cdx2 expression. Cell Death Differ 2017; 24: 819–831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Rubin SJS, Bai L, Haileselassie Y, et al. Mass cytometry reveals systemic and local immune signatures that distinguish inflammatory bowel diseases. Nat Commun 2019; 10: 2686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Guo H, Gibson SA, Ting JPY. Gut microbiota, NLR proteins, and intestinal homeostasis. J Exp Med 2020; 217: e20181832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Mehandru S, Colombel JF. The intestinal barrier, an arbitrator turned provocateur in IBD. Nat Rev Gastroenterol Hepatol 2021; 18: 83–84. [DOI] [PubMed] [Google Scholar]
- 10. Khatri V, Kalyanasundaram R. Therapeutic implications of inflammasome in inflammatory bowel disease. FASEB J 2021; 35: e21439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Peng J, Hu T, Li J, et al. Shepherd’s purse polyphenols exert its anti-inflammatory and antioxidative effects associated with suppressing MAPK and NF-κB pathways and heme oxygenase-1 activation. Oxid Med Cell Longev 2019; 2019: 1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Tao L, Gu X, Xu E, et al. Osthole protects against Ang II-induced endotheliocyte death by targeting NF-κB pathway and Keap-1/Nrf2 pathway. Am J Transl Res 2019; 11: 142–159. [PMC free article] [PubMed] [Google Scholar]
- 13. Seelbinder B, Chen J, Brunke S, et al. Antibiotics create a shift from mutualism to competition in human gut communities with a longer-lasting impact on fungi than bacteria. Microbiome 2020; 8: 133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology 2014; 146: 1489–1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Kayama H, Okumura R, Takeda K. Interaction between the Microbiota, epithelia, and immune cells in the intestine. Annu Rev Immunol 2020; 38: 23–48. [DOI] [PubMed] [Google Scholar]
- 16. Chassaing B, Koren O, Goodrich JK, et al. Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome. Our Nat 2015; 519: 92–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Willems RPJ, van Dijk K, Ket JCF, et al. Evaluation of the association between gastric acid suppression and risk of intestinal colonization with multidrug-resistant microorganisms: a systematic review and meta-analysis. JAMA Intern Med 2020; 180: 561–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Ostaff MJ, Stange EF, Wehkamp J. Antimicrobial peptides and gut microbiota in homeostasis and pathology. EMBO Mol Med 2013; 5: 1465–1483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Zihni C, Mills C, Matter K, et al. Tight junctions: from simple barriers to multifunctional molecular gates. Nat Rev Mol Cell Biol 2016; 17: 564–580. [DOI] [PubMed] [Google Scholar]
- 20. Hartsock A, Nelson WJ. Adherens and tight junctions: structure, function and connections to the actin cytoskeleton. Biochim Biophys Acta 2008; 1778: 660–669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Schneeberger EE, Lynch RD. The tight junction: a multifunctional complex. Am J Physiol Cell Physiol 2004; 286: C1213–C1228. [DOI] [PubMed] [Google Scholar]
- 22. Li Y, Jin L, Chen T. The effects of secretory IgA in the mucosal immune system. Biomed Res Int 2020; 2020: 1–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Colgan SP, Furuta GT, Taylor CT. Hypoxia and innate immunity: keeping up with the HIFsters. Annu Rev Immunol 2020; 38: 341–363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Ren W, Wang K, Yin J, et al. Glutamine-induced secretion of intestinal secretory immunoglobulin A: a mechanistic perspective. Front Immunol 2016; 7: 503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Corthésy B. Role of secretory IgA in infection and maintenance of homeostasis. Autoimmun Rev 2013; 12: 661–665. [DOI] [PubMed] [Google Scholar]
- 26. Chen Y, Cui W, Li X, et al. Interaction between commensal bacteria, immune response and the intestinal barrier in inflammatory bowel disease. Front Immunol 2021; 12: 761981. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Ting JPY, Lovering RC, Alnemri ES, et al. The NLR gene family: a standard nomenclature. Immunity 2008; 28: 285–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Chu JQ, Gao FF, Wu W, et al. Expression profiles of NOD-like receptors and regulation of NLRP3 inflammasome activation in Toxoplasma gondii-infected human small intestinal epithelial cells. Parasit Vectors 2021; 14: 153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Zheng C. The emerging roles of NOD-like receptors in antiviral innate immune signaling pathways. Int J Biol Macromol 2021; 169: 407–413. [DOI] [PubMed] [Google Scholar]
- 30. Kufer TA, Kremmer E, Banks DJ, et al. Role for erbin in bacterial activation of Nod2. Infect Immun 2006; 74: 3115–3124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Hasegawa M, Yamazaki T, Kamada N, et al. Nucleotide-binding oligomerization domain 1 mediates recognition of Clostridium difficile and induces neutrophil recruitment and protection against the pathogen. J Immunol 2011; 186: 4872–4880. [DOI] [PubMed] [Google Scholar]
- 32. Hara H, Seregin SS, Yang D, et al. The NLRP6 inflammasome recognizes lipoteichoic acid and regulates gram-positive pathogen infection. Cell 2018; 175: 1651–1664.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Chen GY. Role of nlrp6 and nlrp12 in the maintenance of intestinal homeostasis. Eur J Immunol 2014; 44: 321–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Mandal S, Rajarammohan S, Kaur J. ROS accumulation and associated cell death mediates susceptibility to Alternaria brassicae in Arabidopsis accessions. Physiol Mol Plant Pathol 2019; 107: 51–59. [Google Scholar]
- 35. Saur IML, Panstruga R, Schulze-Lefert P. NOD-like receptor-mediated plant immunity: from structure to cell death. Nat Rev Immunol 2021; 21: 305–318. [DOI] [PubMed] [Google Scholar]
- 36. Chen GY, Shaw MH and Nunez G. The innate immune receptor Nod1 protects the intestine from inflammation-induced tumorigenesis. Cancer Res 2008; 68: 10060–10067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Le Bourhis L, Magalhaes JG, Selvanantham T, et al. Role of Nod1 in mucosal dendritic cells during Salmonella pathogenicity island 1-independent Salmonella enterica serovar Typhimurium infection. Infect Immun 2009; 77: 4480–4486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Al Nabhani Z, Lepage P, Mauny P, et al. Nod2 deficiency leads to a specific and transmissible mucosa-associated microbial dysbiosis which is independent of the mucosal barrier defect. J Crohns Colitis 2016; 10: 1428–1436. [DOI] [PubMed] [Google Scholar]
- 39. Jiang W, Wang X, Zeng B, et al. Recognition of gut microbiota by NOD2 is essential for the homeostasis of intestinal intraepithelial lymphocytes. J Exp Med 2013; 210: 2465–2476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Richardson WM, Sodhi CP, Russo A. Nucleotide-binding oligomerization domain-2 inhibits toll-like receptor-4 signaling in the intestinal epithelium. Gastroenterology 2010; 139: 904–917.e9176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Shanahan MT, Carroll IM, Grossniklaus E. Mouse Paneth cell antimicrobial function is independent of Nod2. Gut 2014; 63: 903–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Corridoni D, Rodriguez-Palacios A, Di Stefano G, et al. Genetic deletion of the bacterial sensor NOD2 improves murine Crohn’s disease-like ileitis independent of functional dysbiosis. Mucosal Immunol 2017; 10: 971–982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Sellin ME, Muller AA, Hardt WD. Epithelium-intrinsic NAIP/NLRC4 inflammasome drives infected enterocyte expulsion to restrict Salmonella replication in the intestinal mucosa. Cell Host Microbe 2014; 16: 237–248. [DOI] [PubMed] [Google Scholar]
- 44. Franchi L, Kamada N, Nunez G. NLRC4-driven production of IL-1beta discriminates between pathogenic and commensal bacteria and promotes host intestinal defense. Nat Immunol 2012; 13: 449–456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Steiner A, Reygaerts T, Masters SL, et al. Recessive NLRC4-Autoinflammatory Disease Reveals an Ulcerative Colitis Locus. J Clin Immunol 2022; 42: 325–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Williams TM, Leeth RA, Rothschild DE, et al. The NLRP1 inflammasome attenuates colitis and colitis-associated tumorigenesis. J Immunol 2015; 194: 3369–3380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Wang Z, Chen T, Yang C, et al. Secoisolariciresinol diglucoside suppresses Dextran sulfate sodium salt-induced colitis through inhibiting NLRP1 inflammasome. Int Immunopharmacol 2020; 78: 105931. [DOI] [PubMed] [Google Scholar]
- 48. Ranson N, Veldhuis M, Mitchell B. NLRP3-Dependent and -Independent Processing of Interleukin (IL)-1β in Active Ulcerative Colitis. Int J Mol Sci 2018; 20: 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Chen X, Liu G, Yuan L. NEK7 interacts with NLRP3 to modulate the pyroptosis in inflammatory bowel disease via NF-κB signaling. Cell Death Dis 2019; 10: 906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Yao X, Zhang C, Xing Y, et al. Remodelling of the gut microbiota by hyperactive NLRP3 induces regulatory T cells to maintain homeostasis. Nat Commun 2017; 8: 1896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Kempster SL, Belteki G, Forhead AJ. Developmental control of the Nlrp6 inflammasome and a substrate, IL-18, in mammalian intestine. Am J Physiol Gastrointest Liver Physiol. 2011; 300: G253–G263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Yin J, Sheng B, Yang H. The protective roles of NLRP6 in intestinal epithelial cells. Cell Prolif 2019; 52: e12555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Chen L, Wilson JE, Koenigsknecht MJ, et al. NLRP12 attenuates colon inflammation by maintaining colonic microbial diversity and promoting protective commensal bacterial growth. Nat Immunol 2017; 18: 541–551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Kawamoto S, Maruya M, Kato LM, et al. Foxp3(+) T cells regulate immunoglobulin a selection and facilitate diversification of bacterial species responsible for immune homeostasis. Immunity 2014; 41: 152–165. [DOI] [PubMed] [Google Scholar]
- 55. Elias-Oliveira J, Leite JA, Pereira ÍS, et al. NLR and intestinal dysbiosis-associated inflammatory illness: drivers or dampers? Front Immunol 2020; 11: 1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Elinav E, Strowig T, Kau AL, et al. NLRP6 inflammasome regulates colonic microbial ecology and risk for colitis. Cell 2011; 145: 745–757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Seregin SS, Golovchenko N, Schaf B. NLRP6 Protects Il10-/- Mice from Colitis by Limiting Colonization of Akkermansia muciniphila. Cell Rep 2017; 19: 733–745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Anand PK, Malireddi RK, Lukens JR, et al. NLRP6 negatively regulates innate immunity and host defence against bacterial pathogens. Our Nat 2012; 488: 389–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Hirota SA, Ng J, Lueng A, et al. The NLRP3 inflammasome plays key role in the regulation of intestinal homeostasis. Inflamm Bowel Dis 2011; 17: 1359–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Bauer C, Duewell P, Mayer C, et al. Colitis induced in mice with dextran sulfate sodium (DSS) is mediated by the NLRP3 inflammasome. Gut 2010; 59: 1192–1199. [DOI] [PubMed] [Google Scholar]
- 61. Liu Z, Zaki MH, Vogel P, et al. Role of inflammasomes in host defense against Citrobacter rodentium infection. J Biol Chem 2012; 287: 16955–16964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Yoganathan P, Rossel JB, Jordi SBU, et al.; Swiss IBD cohort study group. Genotype-phenotype associations of polymorphisms within the gene locus of NOD-like receptor pyrin domain containing 3 in Swiss inflammatory bowel disease patients. BMC Gastroenterol 2021; 21: 310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Singh V, Yeoh BS, Walker RE, et al. Microbiota fermentation-NLRP3 axis shapes the impact of dietary fibres on intestinal inflammation. Gut 2019; 68: 1801–1812. [DOI] [PubMed] [Google Scholar]
- 64. Bauer C, Duewell P, Lehr HA, et al. Protective and aggravating effects of nlrp3 inflammasome activation in IBD models: influence of genetic and environmental factors. Dig Dis 2012; 30 Suppl 1: 82–90. [DOI] [PubMed] [Google Scholar]
- 65. Biswas A, Kobayashi KS. Regulation of intestinal microbiota by the NLR protein family. Int Immunol 2013; 25: 207–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Sun X, Jobin C. 890 defective NOD2-Induced bactericidal activity exacerbates Campylobacter Jejuni-Induced Colitis in Il10-/- mice. Gastroenterol 2013; 144: S–157. [Google Scholar]
- 67. Couturier-Maillard A, Secher T, Rehman A, et al. NOD2-mediated dysbiosis predisposes mice to transmissible colitis and colorectal cancer. J Clin Investig 2013; 123: 700–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Zaki MH, Vogel P, Malireddi RK, et al. The NOD-like receptor NLRP12 attenuates colon inflammation and tumorigenesis. Cancer Cell 2011; 20: 649–660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Frank DN, St Amand AL, Feldman RA, et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci USA 2007; 104: 13780–13785. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Schubert AM, Rogers MA, Ring C, et al. Microbiome data distinguish patients with Clostridium difficile infection and non-C. Difficile-associated diarrhea from healthy controls. mBio 2014; 5: e01021–NaN14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Haberman Y, Tickle TL, Dexheimer PJ, et al. Pediatric Crohn disease patients exhibit specific ileal transcriptome and microbiome signature. J Clin Investig 2014; 124: 3617–3633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Shen L, Su L, Turner JR. Mechanisms and functional implications of intestinal barrier defects. Dig Dis 2009; 27: 443–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Chelakkot C, Ghim J, Ryu SH. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp Mol Med 2018; 50: 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Miyoshi J, Takai Y. Structural and functional associations of apical junctions with cytoskeleton. Biochim et Biophys Acta 2008; 1778: 670–691. [DOI] [PubMed] [Google Scholar]
- 75. Pinton P, Nougayrède JP, Del Rio JC, et al. The food contaminant deoxynivalenol, decreases intestinal barrier permeability and reduces claudin expression. Toxicol Appl Pharmacol 2009; 237: 41–48. [DOI] [PubMed] [Google Scholar]
- 76. Su L, Nalle SC, Shen L, et al. TNFR2 activates MLCK-dependent tight junction dysregulation to cause apoptosis-mediated barrier loss and experimental colitis. Gastroenterol 2013; 145: 407–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Al Nabhani Z, Montcuquet N, Roy M, et al. Complementary roles of nod2 in hematopoietic and nonhematopoietic cells in preventing gut barrier dysfunction dependent on MLCK activity. Inflamm Bowel Dis 2017; 23: 1109–1119. [DOI] [PubMed] [Google Scholar]
- 78. Feng Y, Wang Y, Wang P, et al. Short-chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell Physiol Biochem 2018; 49: 190–205. [DOI] [PubMed] [Google Scholar]
- 79. Feng Y, Huang Y, Wang Y, et al. Antibiotics induced intestinal tight junction barrier dysfunction is associated with microbiota dysbiosis, activated NLRP3 inflammasome and autophagy. PLoS One 2019; 14: e0218384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Du J, Chen Y, Shi Y, et al. 1,25-dihydroxyvitamin D protects intestinal epithelial barrier by regulating the myosin light chain kinase signaling pathway. Inflamm Bowel Dis 2015; 21: 2495–2506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Wlodarska M, Thaiss CA, Nowarski R, et al. NLRP6 inflammasome orchestrates the colonic host-microbial interface by regulating goblet cell mucus secretion. Cell 2014; 156: 1045–1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Normand S, Delanoye-Crespin A, Bressenot A, et al. Nod-like receptor pyrin domain-containing protein 6 (NLRP6) controls epithelial self-renewal and colorectal carcinogenesis upon injury. Proc Natl Acad Sci USA 2011; 108: 9601–9606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Zaki MH, Boyd KL, Vogel P, et al. The NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis. Immun 2010; 32: 379–391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Song-Zhao GX, Srinivasan N, Pott J, et al. Nlrp3 activation in the intestinal epithelium protects against a mucosal pathogen. Mucosal Immunol 2014; 7: 763–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Zhang P, Liu Y, Hu L, et al. NLRC4 inflammasome-dependent cell death occurs by a complementary series of three death pathways and determines lethality in mice. Sci Adv 2021; 7: eabi9471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Cummings JR, Cooney RM, Clarke G, et al. The genetics of NOD-like receptors in Crohn's disease. Tissue Antigens 2010; 76: 48–56. [DOI] [PubMed] [Google Scholar]
- 87. Jamontt J, Petit S, Clark N, et al. Nucleotide-binding oligomerization domain 2 signaling promotes hyperresponsive macrophages and colitis in IL-10-deficient mice. J Immunol 2013; 190: 2948–2958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Nayar S, Morrison JK, Giri M, et al. A myeloid-stromal niche and gp130 rescue in NOD2-driven Crohn’s disease. Our Nat 2021; 593: 275–281 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Cui S, Wang C, Bai W. CD1d1 intrinsic signaling in macrophages controls NLRP3 inflammasome expression during inflammation. Sci Adv 2020; 6: eaaz7290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Sugimoto K, Ogawa A, Mizoguchi E, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Investig 2008; 118: 534–544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Bishop JL, Roberts ME, Beer JL, et al. Lyn activity protects mice from DSS colitis and regulates the production of IL-22 from innate lymphoid cells. Mucosal Immunol 2014; 7: 405–416. [DOI] [PubMed] [Google Scholar]
- 92. Chen GY, Liu M, Wang F, et al. A functional role for nlrp6 in intestinal inflammation and tumorigenesis. J Immunol 2011; 186: 7187–7194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Chavan SS, Pavlov VA, Tracey KJ. Mechanisms and therapeutic relevance of neuro-immune Communication. Immun 2017; 46: 927–942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Chaubey P, Momin M, Sawarkar S. Significance of ligand-anchored polymers for drug targeting in the treatment of colonic disorders. Front Pharmacol 2019; 10: 1628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Ferrusquía J, Pérez-Martínez I, Gómez de la Torre R, et al. Gastroenterology case report of mesalazine-induced cardiopulmonary hypersensitivity. World J Gastroenterol 2015; 21: 4069–4077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Leung B, Lowery D. The patent landscape of inflammasome modulators. Nat Rev Drug Discov 2020; 19: 158. [DOI] [PubMed] [Google Scholar]
- 97. Schwaid AG, Spencer KB. Strategies for targeting the NLRP3 inflammasome in the clinical and preclinical space. J Med Chem 2021; 64: 101–122. [DOI] [PubMed] [Google Scholar]
- 98. Sun Y, Zhao Y, Yao J, et al. Wogonoside protects against dextran sulfate sodium-induced experimental colitis in mice by inhibiting NF-κB and NLRP3 inflammasome activation. Biochem Pharmacol 2015; 94: 142–154. [DOI] [PubMed] [Google Scholar]
- 99. Zhao Y, Guo Q, Zhu Q, et al. Flavonoid VI-16 protects against DSS-induced colitis by inhibiting Txnip-dependent NLRP3 inflammasome activation in macrophages via reducing oxidative stress. Mucosal Immunol 2019; 12: 1150–1163. [DOI] [PubMed] [Google Scholar]
- 100. Gong Z, Zhao S, Zhou J, et al. Curcumin alleviates DSS-induced colitis via inhibiting NLRP3 inflammsome activation and IL-1β production. Mol Immunol 2018; 104: 11–19. [DOI] [PubMed] [Google Scholar]
- 101. Chen L, You Q, Hu L, et al. The antioxidant procyanidin reduces reactive oxygen species signaling in macrophages and ameliorates experimental colitis in mice. Front Immunol 2017; 8: 1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Cao H, Liu J, Shen P, et al. Protective Effect of naringin on DSS-Induced ulcerative colitis in mice. J Agric Food Chem 2018; 66: 13133–13140. [DOI] [PubMed] [Google Scholar]
- 103. Zhang Z, Li S, Cao H, et al. The protective role of phloretin against dextran sulfate sodium-induced ulcerative colitis in mice. Food Funct 2019; 10: 422–431. [DOI] [PubMed] [Google Scholar]
- 104. Radulovic K, Normand S, Rehman A, et al. A dietary flavone confers communicable protection against colitis through NLRP6 signaling independently of inflammasome activation. Mucosal Immunol 2018; 11: 811–819. [DOI] [PubMed] [Google Scholar]
- 105. Wang K, Lv Q, Miao YM, et al. Cardamonin, a natural flavone, alleviates inflammatory bowel disease by the inhibition of NLRP3 inflammasome activation via an AhR/Nrf2/NQO1 pathway. Biochem Pharmacol 2018; 155: 494–509. [DOI] [PubMed] [Google Scholar]
- 106. He X, Wei Z, Wang J, et al. Alpinetin attenuates inflammatory responses by suppressing TLR4 and NLRP3 signaling pathways in DSS-induced acute colitis. Sci Rep 2016; 6: 28370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Liu G, Zhao W, Bai J, et al. Formononetin protects against concanavalin-A-induced autoimmune hepatitis in mice through its anti-apoptotic and anti-inflammatory properties. Biochem Cell Biol 2021; 99: 231–240. [DOI] [PubMed] [Google Scholar]
- 108. Chen Y, Le TH, Du Q, et al. Genistein protects against DSS-induced colitis by inhibiting NLRP3 inflammasome via TGR5-CAMP signaling. Int Immunopharmacol 2019; 71: 144–154. [DOI] [PubMed] [Google Scholar]
- 109. Luo X, Wang X, Huang S, et al. Paeoniflorin ameliorates experimental colitis by inhibiting gram-positive bacteria-dependent MDP-NOD2 pathway. Int Immunopharmacol 2021; 90: 107224. [DOI] [PubMed] [Google Scholar]
- 110. Fan H, Chen W, Zhu J, et al. Toosendanin alleviates dextran sulfate sodium-induced colitis by inhibiting M1 macrophage polarization and regulating NLRP3 inflammasome and Nrf2/ho-1 signaling. Int Immunopharmacol 2019; 76: 105909. [DOI] [PubMed] [Google Scholar]
- 111. Guo W, Liu W, Jin B, et al. Asiatic acid ameliorates dextran sulfate sodium-induced murine experimental colitis via suppressing mitochondria-mediated NLRP3 inflammasome activation. Int Immunopharmacol 2015; 24: 232–238. [DOI] [PubMed] [Google Scholar]
- 112. Liang R, Chen W, Fan H, et al. Dihydroartemisinin prevents dextran sodium sulphate-induced colitisthrough inhibition of the activation of NLRP3 inflammasome and p38 MAPK signaling. Int Immunopharmacol 2020; 88: 106949. [DOI] [PubMed] [Google Scholar]
- 113. Zhu G, Wang H, Wang T, et al. Ginsenoside Rg1 attenuates the inflammatory response in DSS-induced mice colitis. Int Immunopharmacol 2017; 50: 1–5. [DOI] [PubMed] [Google Scholar]
- 114. Saber S, Abd El-Kader EM, Sharaf H, et al. Celastrol augments sensitivity of NLRP3 to CP-456773 by modulating HSP-90 and inducing autophagy in dextran sodium sulphate-induced colitis in rats. Toxicol Appl Pharmacol 2020; 400: 115075. [DOI] [PubMed] [Google Scholar]
- 115. Fan M, Xiang G, Chen J, et al. Libertellenone M, a diterpene derived from an endophytic fungus Phomopsis sp. S12, protects against DSS-induced colitis via inhibiting both nuclear translocation of NF-κB and NLRP3 inflammasome activation. Int Immunopharmacol 2020; 80: 106144. [DOI] [PubMed] [Google Scholar]
- 116. Tian M, Ma P, Zhang Y, et al. Ginsenoside Rk3 alleviated DSS-induced ulcerative colitis by protecting colon barrier and inhibiting NLRP3 inflammasome pathway. Int Immunopharmacol 2020; 85: 106645. [DOI] [PubMed] [Google Scholar]
- 117. Mai CT, Wu MM, Wang CL, et al. Palmatine attenuated dextran sulfate sodium (DSS)-induced colitis via promoting mitophagy-mediated NLRP3 inflammasome inactivation. Mol Immunol 2019; 105: 76–85. [DOI] [PubMed] [Google Scholar]
- 118. Guo W, Hu S, Elgehama A, et al. Fumigaclavine C ameliorates dextran sulfate sodium-induced murine experimental colitis via NLRP3 inflammasome inhibition. J Pharmacol Sci 2015; 129: 101–106. [DOI] [PubMed] [Google Scholar]
- 119. Shen P, Zhang Z, Zhu K, et al. Evodiamine prevents dextran sulfate sodium-induced murine experimental colitis via the regulation of NF-kappaB and NLRP3 inflammasome, Biomed Pharmacother 2019; 110:786-795. [DOI] [PubMed] [Google Scholar]
- 120. Du X, Chen W, Wang Y, et al. Therapeutic efficacy of carboxyamidotriazole on 2,4,6-trinitrobenzene sulfonic acid-induced colitis model is associated with the inhibition of NLRP3 inflammasome and NF-κB activation. Int Immunopharmacol 2017; 45: 16–25. [DOI] [PubMed] [Google Scholar]
- 121. Wang Y, Wang H, Qian C, et al. 3-(2-Oxo-2-phenylethylidene)-2,3,6,7-tetrahydro-1H-pyrazino[2,1-a]isoquinolin-4(11bH)-one (compound 1), a novel potent Nrf2/are inducer, protects against DSS-induced colitis via inhibiting NLRP3 inflammasome. Biochem Pharmacol 2016; 101: 71–86. [DOI] [PubMed] [Google Scholar]
- 122. Zeng J, Zhang D, Wan X, et al. Chlorogenic acid suppresses miR-155 and ameliorates ulcerative colitis through the NF-κB/NLRP3 inflammasome pathway. Mol Nutr Food Res 2020; 64(23):e2000452. [DOI] [PubMed] [Google Scholar]
- 123. Lopes de Oliveira GA, Alarcón de la Lastra C, Rosillo MÁ, et al. Preventive effect of bergenin against the development of TNBS-induced acute colitis in rats is associated with inflammatory mediators inhibition and NLRP3/ASC inflammasome signaling pathways. Chem Biol Interact 2019; 297: 25–33. [DOI] [PubMed] [Google Scholar]
- 124. Zhou J, Wang T, Dou Y, et al. Brusatol ameliorates 2, 4, 6-trinitrobenzenesulfonic acid-induced experimental colitis in rats: involvement of NF-κB pathway and NLRP3 inflammasome. Int Immunopharmacol 2018; 64: 264–274. [DOI] [PubMed] [Google Scholar]
- 125. Huang Y, Li W, Kong AN. Anti-oxidative stress regulator NF-E2-related factor 2 mediates the adaptive induction of antioxidant and detoxifying enzymes by lipid peroxidation metabolite 4-hydroxynonenal. Cell Biosci 2012; 2: 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Wu XF, Ouyang ZJ, Feng LL, et al. Suppression of NF-κB signaling and NLRP3 inflammasome activation in macrophages is responsible for the amelioration of experimental murine colitis by the natural compound fraxinellone. Toxicol Appl Pharmacol 2014; 281: 146–156. [DOI] [PubMed] [Google Scholar]
- 127. Cao R, Ma Y, Li S, et al. 1,25(OH)2 D3 alleviates DSS-induced ulcerative colitis via inhibiting NLRP3 inflammasome activation. J Leukoc Biol 2020; 108: 283–295. [DOI] [PubMed] [Google Scholar]
- 128. Jiang X, Zhong L, Sun D, et al. Magnesium lithospermate B acts against dextran sodiumsulfate-induced ulcerative colitis by inhibiting activation of the NRLP3/ASC/Caspase-1 pathway. Environ Toxicol Pharmacol 2016; 41: 72–77. [DOI] [PubMed] [Google Scholar]
- 129. Zhang Q, Xu N, Hu X, et al. Anti-colitic effects of physalin B on dextran sodium sulfate-induced BALB/c mice by suppressing multiple inflammatory signaling pathways. J Ethnopharmacol 2020; 259: 112956. [DOI] [PubMed] [Google Scholar]
- 130. Xia X, Lin H, Luo F, et al. Oryzanol ameliorates DSS-Stimulated gut barrier damage via targeting the gut Microbiota accompanied by the TLR4/NF-κB/NLRP3 cascade response in vivo. J Agric Food Chem 2022; 70: 15747–15762. [DOI] [PubMed] [Google Scholar]
- 131. Ma X, Di Q, Li X, et al. Munronoid I ameliorates DSS-Induced mouse colitis by inhibiting NLRP3 inflammasome activation and pyroptosis via modulation of NLRP3. Front Immunol 2022; 13: 853194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Shao M, Yan Y, Zhu F, et al. Artemisinin analog SM934 alleviates epithelial barrier dysfunction via inhibiting apoptosis and caspase-1-mediated pyroptosis in experimental colitis. Front Pharmacol 2022; 13: 849014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Gao X, Fan W, Tan L, et al. Soy isoflavones ameliorate experimental colitis by targeting ERα/NLRP3 inflammasome pathways. J Nutr Biochem 2020; 83: 108438. [DOI] [PubMed] [Google Scholar]
- 134. Zong S, Ye Z, Zhang X, et al. Protective effect of Lachnum polysaccharide on dextran sulfate sodium-induced colitis in mice. Food Funct 2020; 11: 846–859. [DOI] [PubMed] [Google Scholar]
- 135. Sun Z, Pei W, Guo Y, et al. Gut microbiota-mediated NLRP12 expression drives the attenuation of dextran sulphate sodium-induced ulcerative colitis by Qingchang Wenzhong decoction. Evid Based Complement Alternat Med 2019; 2019: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Zhang J, Kang X, Sun M, et al. Qingre Jianpi decoction attenuates inflammatory responses by suppressing NOD-like receptor family pyrin domain-containing 3 inflammasome activation in dextran sulfate sodium-induced colitis mice. J Tradit Chin Med 2021; 41: 68–78. [DOI] [PubMed] [Google Scholar]
- 137. Tian Z, Liu Y, Yang B, et al. Astagalus polysaccharide attenuates murine colitis through inhibiton of the NLRP3 inflammasome. Planta Med 2017; 83: 70–77. [DOI] [PubMed] [Google Scholar]
- 138. Zong S, Yang L, Park HJ, et al. Dietary intake of Lycium ruthenicum Murray ethanol extract inhibits colonic inflammation in dextran sulfate sodium-induced murine experimental colitis. Food Funct 2020; 11: 2924–2937. [DOI] [PubMed] [Google Scholar]
- 139. Saba E, Lee YY, Kim M, et al. A novel herbal formulation consisting of red ginseng extract and Epimedium koreanum Nakai-attenuated dextran sulfate sodium-induced colitis in mice. J Ginseng Res 2020; 44: 833–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Mahmoud TN, El-Maadawy WH, Kandil ZA, et al. Canna x generalis L.H. Bailey rhizome extract ameliorates dextran sulfate sodium-induced colitis via modulating intestinal mucosal dysfunction, oxidative stress, inflammation, and TLR4/ NF-κs and NLRP3 inflammasome pathways. J Ethnopharmacol 2021; 269: 113670. [DOI] [PubMed] [Google Scholar]
- 141. Wang Z, Li C, He X, et al. Platycodon grandiflorum root fermentation broth reduces inflammation in a mouse IBD model through the AMPK/NF-κB/NLRP3 pathway. Food Funct 2022; 13: 3946–3956. [DOI] [PubMed] [Google Scholar]



