Next Article in Journal
Maternal, Perinatal and Neonatal Outcomes of Triplet Pregnancies According to Chorionicity: A Systematic Review of the Literature and Meta-Analysis
Next Article in Special Issue
Management of Preschool Wheezing: Guideline from the Emilia-Romagna Asthma (ERA) Study Group
Previous Article in Journal
Critically-Ill Patients with Biliary Obstruction and Cholangitis: Bedside Fluoroscopic-Free Endoscopic Drainage versus Percutaneous Drainage
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

NLRP3, NLRC4 and NLRC5 Gene Polymorphisms Associate with Susceptibility of Pulmonary Aspergillosis in Non-Neutropenic Patients

Department of Respiratory and Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Clin. Med. 2022, 11(7), 1870; https://doi.org/10.3390/jcm11071870
Submission received: 25 February 2022 / Revised: 22 March 2022 / Accepted: 25 March 2022 / Published: 28 March 2022
(This article belongs to the Special Issue Clinical Management of Respiratory Tract Infection)

Abstract

:
Background: Non-neutropenic pulmonary aspergillosis is one of the most common and serious fungal infections. Previous studies have shown that single nucleotide polymorphisms (SNPs) of pattern recognition receptors genes are associated with susceptibility to aspergillosis. NOD-like receptors (NLRs) play an important role in the immunological response against fungal infection. In this study, we investigated the relationship between polymorphisms of three NLRs and susceptibility to pulmonary aspergillosis disease in non-neutropenic patients. Methods: We included 73 patients with proven pulmonary aspergillosis and 103 healthy controls. A total of sixteen SNPs in the NLRP3, NLRC4, and NLRC5 genes were detected by PCR-direct sequencing. Then, we evaluated the association between these polymorphisms and susceptibility to aspergillosis. Results: Fifteen SNPs were consistent with Hardy–Weinberg equilibrium except for NLRP3 rs7525979. A total of eight SNPs (NLRP3 rs3806265, NLRC4 rs212704 and NLRC5 rs1684579, rs12598522, rs3995817, rs3995818, rs34531240, rs28438857) were observed an association with susceptibility of pulmonary aspergillosis. The CC homozygote of NLRP3 rs3806265, TT homozygote of NLRC5 rs1684579 and T allele of NLRC5 rs12598522 were associated with a higher risk of aspergillosis while TT homozygote of NLRC4 rs212704 was associated with a lower risk of aspergillosis. Especially in the invasive pulmonary aspergillosis subgroup, the TT homozygote of NLRC5 rs1684579 and rs3995817, the CC homozygote of NLRC5 rs34531240 and rs28438857, GG homozygote of NLRC5 rs3995818, the C allele and CC homozygote of NLRP3 rs3806265 were associated with higher susceptibility. Conclusions: This study showed an association between polymorphisms of NLRP3, NLRC4, and NLRC5 and susceptibility to pulmonary aspergillosis for the first time. Further investigations in larger populations are needed, and functional studies are also required to investigate the function of these NLRs in aspergillosis, as well as other fungal infection diseases.

1. Introduction

Aspergillus is a saprotrophic fungus which spreads by asexual conidia. The airborne Aspergillus conidia can be inhaled into the respiratory tract and lungs of humans, causing different types of diseases, including invasive pulmonary aspergillosis (IPA), chronic pulmonary aspergillosis (CPA), and allergic bronchial pulmonary aspergillosis (ABPA) [1,2,3]. Since the late 1990s, aspergillosis has actually proved to be the most common invasive pulmonary fungal infection. Moreover, it has also become the most expensive fungal disease because of its prevalence and costly treatments [4].
Pattern recognition receptors (PRRs), including Toll-like receptors, RIG-I-like receptors, NOD-like receptors, and C-type lectin receptors, play an important role in host immunity against Aspergillus; thus, the genetic defects in PRRs may lead to susceptibility to aspergillosis [5,6,7]. Polymorphisms in Toll-like receptors such as TLR4, C-type lectin receptors such as dectin-1, and other pattern recognition receptors such as Pentraxin 3 (PTX3) have been found to be associated with susceptibility to aspergillosis [8,9,10].
The NOD-like receptors make up an important family of PRRs. Many of them can bind with apoptosis-associated speck-like protein containing a CARD(ASC) and caspase-1 to form inflammasome, such as NLRP3, NLRC4, NLRC5 inflammasome, and so on [11]. In an aspergillosis mouse model, several kinds of NLRs increased in the infected lungs, including NLRP3, NLRC4, and NLRC5, but their exact functions remain to be explored [12]. NLRP3 is the most well-characterized and most well-studied inflammasome sensor molecule. When the NLRP3 inflammasome is activated, Pro-Caspase-1 is cleaved, which leads to the release of proinflammatory cytokines like IL-1β and IL-18, as well as pyroptotic cell death [13,14]. Generally, NLRP3 is known to contribute to antifungal immunity and help control infection [5]. For instance, a GAG-deficient Aspergillus mutant, which failed to elicit protective NLRP3 inflammasome activation, exhibited enhanced virulence [15]. Likewise, the mice lacking AIM2 and NLRP3 were susceptible to Aspergillus infection [16]. Similarly, the NLRC4 inflammasome has been also found to protect mucosal barriers such as the lung, stomach, and intestine from invading pathogens [17]. NLRC5 is the largest one in the NLR family and works as the master transcriptional regulator of MHC class I and related genes [18,19]. Therefore, after infection, NLRC5 knockout mice showed increased bacterial load and impaired clearance of viruses due to strongly impaired MHCI-mediated CD8+ T cell activation [20,21,22]. However, the role of NLRC4 and NLRC5 in Aspergillus infection is still unknown. Since innate immunity and adaptive immune responses are all important parts for host defense against Aspergillus, we assume that they are probably involved in the host immune response in aspergillosis, as well as in other PRRs.
A few previous studies have shown the polymorphisms of NLRs genes affected the susceptibility of Aspergillus infection or colonization. Among transplant recipients after hematopoietic stem cell transplantation (HSCT), P268S (rs2066842) in NOD2 of the donors was associated with an increased risk of invasive aspergillosis [23]. For patients with cystic fibrosis, significant associations were found between A.fumigatus colonization and polymorphisms of NLRC4, including the haplotype ACTT (rs212704 rs455060 rs7562653 rs385076) and GG genotype of rs212704 [24]. In this study, we investigated 16 SNPs in NLRP3, NLRC4, and NLRC5 genes among the southeastern Han Chinese population and analyzed the relationships between these SNPs and susceptibility of pulmonary aspergillosis in non-neutropenic patients.

2. Materials and Methods

2.1. Study Population

This study included 73 pulmonary aspergillosis patients treated at Jinling Hospital from June 2016 to December 2019. The control group consisted of 103 healthy people undergoing physical examination. According to the updated IDSA guideline criteria and EORTC/MSGERC criteria, these aspergillosis patients were diagnosed as 30 IPA, 27 CPA (including chronic cavitary pulmonary aspergillosis, CCPA and aspergilloma), and 16 ABPA [25,26] (Table 1). Since the study subjects were non-neutropenia patients, we excluded patients who had previously undergone organ transplantation or chemotherapy.

2.2. Selection of SNPs and Genotyping

The single nucleotide polymorphisms of NLRP3 (rs3806265, rs7525979, rs35829419, rs10754558) and NLRC4 (rs12989936, rs212704, rs7562653, rs479333, rs385076) were selected from previous literature. Seven SNPs of NLRC5 (rs12598522, rs34531240, rs28438857, rs3995818, rs3995817, rs1684579, rs3751705) were selected based on information from the NCBI GenBank, dbSNP, and HapMap databases, with the minimum allele frequency set at 5% and r2 at 0.8. These SNPs were located within the coding region, 5′ untranslated region (UTR), or 3′UTR that may possibly influence protein synthesis and gene transcription. Peripheral blood (1 mL) was collected in an EDTA tube from each subject.
Genomic DNA was extracted from the whole blood using the QIAamp DNA Blood Mini Kit (Qiagen, Berlin, Germany) according to the manufacturer’s instructions and then stored in a −80 °C freezer. Primers were designed using Primer Premier 5.0 (Premier Biosoft International, Palo Alto, CA, USA). PCR amplification was performed in Eppendorf PRO PCR System (Hamburg, Germany). All SNPs were genotyped by ABI Prism 377 Sequence Detection System (Applied Biosystems, Foster City, CA, USA) with technical support from the Shanghai Genesky Biotechnology Company (Shanghai, China). DNA sequences were read by Chromas 2.3 software (Technelysium Pty Ltd., Tewantin, Australia). Negative controls were included in each plate for accuracy.

2.3. Statistics

Each SNP was tested for Hardy–Weinberg equilibrium (HWE) in the healthy control group using the chi-square (χ2) test. Statistical comparison was performed by independent Student’s t-test or one-way analysis of variance. Differences among SNPs were evaluated using Pearson’s χ2 or Fisher’s exact test. The strength of association between polymorphisms and the risk of aspergillosis was evaluated by odds ratio (OR) and 95% confidence interval (CI). HWE, linkage disequilibrium, and haplotype analysis were analyzed by HaploView [27,28]. Statistical analysis was performed by the SPSS 24.0 and the GraphPad Prism 7. All tests were considered significant with a p value of <0.05.

3. Results

3.1. Characteristics of the Patient Population

We enrolled 73 patients with pulmonary aspergillosis and 103 healthy controls. Healthy controls were used to compare the frequency of mutations between the patient group and the general population, and there were no significant differences in age (58.48 ± 1.63 vs. 55.25 ± 1.038, p = 0.0819) or sex (male/female: 41/32 vs. 64/39, p = 0.4263) between the case and control groups. Table 1 summarizes the main characteristics of the study population. The case group included 20 (27.40%) patients with hypertension and 12 (16.44%) with diabetes, 11 (15.07%) with COPD, 5 (6.85%) with asthma, 23 (31.51%) with bronchiectasis, and 20 (27.40%) with tuberculosis. In addition, 21 (28.77%) in the case group had a history of smoking and 13 (17.81%) had a history of steroid treatment prior to onset.

3.2. Hardy-Weinberg Equilibrium Analysis and Minor Allele Frequencies of SNPs

We analyzed HWE and minor allele frequencies (MAF) of the 16 SNPs. All but NLRP3 rs7525979 were consistent with HWE in the control group (p > 0.05). No allele was detected at NLRP3 rs35829419 in either control group or case group. MAF of the other SNPs were >5%. So, we excluded these two SNPs in the following statistical analysis. Other information regarding these SNPs is shown in Table 2.

3.3. Association of NLRs Variants with Pulmonary Aspergillosis

First, we evaluated the association of 14 single nucleotide polymorphisms with pulmonary aspergillosis risk among all patients and healthy controls (Table 3). A total of 4 SNPs (NLRP3 rs3806265, NLRC4 rs212704, NLRC5 rs1684579, and rs12598522) were observed an association with aspergillosis risk. Genotype difference in NLRP3 rs3806265 between the case and control groups was statistically significant (p = 0.0451) and the CC homozygote of rs3806265 was associated with a higher risk of aspergillosis (p = 0.0130; OR = 2.567, 95% CI: 1.239 to 5.255). For the NLRC4 rs212704, the TT homozygote was associated with a lower risk of aspergillosis (p = 0.0447; OR = 0.4468, 95%CI: 0.2071 to 0.959) while for the NLRC5 rs1684579 it was opposite (p = 0.0261; OR = 2.066, 95%CI: 1.085 to 4.018). Furthermore, the T allele of NLRC5 rs12598522 was more frequent in aspergillosis patients than healthy controls (p = 0.0305; OR = 1.601, 95%CI: 1.048 to 2.47). The other SNPs did not show any association with aspergillosis risk (Supplementary Table S1).

3.4. Association of NLRs Variants with Different Kinds of Aspergillosis

Next, we divided the patients into non-ABPA subgroup and ABPA subgroup according to whether the pathological process was mainly an inflammatory or allergic response and then compared each of the two subgroups with healthy controls (Table 4). In the non-ABPA group, gene polymorphism of the rs3806265 in NLRP3 and rs12598522, rs1684579 in NLRC5 showed a significant association with susceptibility. Similar to the entire group analysis, genotype difference in NLRP3 rs3806265 was statistically significant (p = 0.0298) and the CC homozygote was associated with a higher risk (p = 0.0129; OR = 2.702, 95%CI: 1.172 to 5.924), as well as the T allele of NLRC5 rs12598522 (p = 0.0419; OR = 1.612, 95%CI: 1.012 to 2.529). For the NLRC5 rs1684579, there was also a significant genotype difference (p = 0.0125) and the TT homozygote (p = 0.0044; OR = 2.665, 95%CI: 1.371 to 5.292), as well as T allele (p = 0.0471; OR = 1.614, 95%CI: 0.9938 to 2.624) were significantly associated with a higher risk of Aspergillus infection. However, in the ABPA subgroup, there was no SNP associated with susceptibility (Supplementary Table S2).
Finally, we divided the non-ABPA group into IPA subgroup and CPA subgroup and compared them with control group (Table 5). It was worth noting that we found rs3806265 in NLRP3 and rs34531240, rs28438857, rs3995818, rs3995817, rs1684579 in NLRC5 were all associated with IPA risk. The CC homozygote (p = 0.0004; OR = 4.861, 95%CI: 2.007 to 11.9) and C allele (p = 0.0117; OR = 2.115, 95%CI: 1.196 to 3.741) of NLRP3 rs3806265, as well as TT homozygote of NLRC5 rs1684579 (p = 0.0036; OR = 3.385, 95%CI: 1.47 to 8.101) were associated with a higher risk of IPA and the genotype differences of them were significant between IPA patients and controls (p = 0.0017; p = 0092). The CC homozygote of rs34531240 and rs28438857, GG homozygote of rs3995818, TT homozygote of rs3995817 in NLRC5 were all more frequent in IPA patients than controls (p = 0.0420; OR = 2.386, 95%CI: 0.9749 to 5.567). However, in the ABPA subgroup, there was no SNP associated with susceptibility (Supplementary Table S3).

3.5. Linkage Disequilibrium and Haplotype Analyses

Linkage disequilibrium (LD) analysis showed that rs3806265, rs7525979 in NLRP3, rs385076, rs47933, rs7562653 in NLRC4, rs34531240, rs28438857, rs3995817, rs3995818 in NLRC5 were in high LD (D′ > 95) (Figure 1). In the NLRP3 rs3806265, rs7525979 block, haplotype TC showed an association with a decreased risk of IPA (p = 0.0117) while haplotype CC was more frequent among IPA patients (p = 0.0136) (Table 6).

4. Discussion

The aim of this study was to investigate the relationship between NLRP3, NLRC4, and NLRC5 gene polymorphisms and susceptibility to pulmonary aspergillosis in non-neutropenic patients among the Chinese population. For the first time, we found rs3806265 in NLRP3, rs212704 in NLRC4 and rs12598522, rs34531240, rs28438857, rs3995818, rs3995817, rs1684579 in NLRC5 were associated with pulmonary aspergillosis in non-neutropenic patients.
The occurrence and development of aspergillosis are closely related to the host’s immune status. Therefore, polymorphisms of many immune-related genes are associated with susceptibility to aspergillosis, such as tumor necrosis factor receptor 1 (TNFR1), TLR1/4/5/6, Dectin-1, DC-SIGN, IL-8, IL-10, IL-12, IL-4R, IFN-γ, IRF4 and so on [8,9,10,29,30,31,32,33,34,35]. Decades ago, pulmonary aspergillosis (PA) often occurred in immunocompromised patients, in particular among hematopoietic stem cell transplants (HSCT) and patients with hematological malignancies. However, there is a rising incidence of pulmonary aspergillosis in non-neutropenic patients during recent years [36,37]. These previous studies were mostly based on patients with severe immunodeficiency, and there were few studies focusing on aspergillosis patients with non-severe immune deficiency, so our study selected non-neutropenic patients as the research object.
NLR proteins are central mediators of microbial sensing with diverse functions, and they play an important role in the host antimicrobial immune responses including anti-Aspergillus response [38]. NLRs are usually composed of a tripartite structure, including an N-terminal effector domain, a central NACHT domain containing the nucleotide binding domain (NBD) for self-oligomerization and C-terminal leucine-rich repeats (LRRs) for recognizing PAMPs or DAMPs [39]. As for NLRs, previous studies have shown that rs2066842 in NOD2 of donors was associated with the risk of invasive aspergillosis after hematopoietic stem-cell transplantation, while rs212704 and ACTT (rs212704 rs455060 rs7562653 rs385076) in NLRC4 were associated with A. fumigatus colonization in cystic fibrosis patients [23,24]. In our study, we found the TT homozygote of rs212704 in NLRC4 and C allele of rs12598522 in NLRC5 was associated with a lower risk of aspergillosis. On the contrary, CC homozygote of rs3806265 in NLRP3 and TT homozygote of rs1684579 in NLRC5 was associated with a high risk of aspergillosis, especially of IPA. Besides, in the IPA subgroup, the CC homozygote of rs34531240 and rs28438857, GG homozygote of rs3995818, TT homozygote of rs3995817 in NLRC5 was more frequent than controls.
The polymorphism of rs3806265 in NLRP3 is associated with several kinds of diseases, such as myasthenia gravis (MG), psoriasis, recurrent aphthous stomatitis (RAS) and relapsing–remitting multiple sclerosis (RRMS) [40,41,42,43]. In the Iranian population, the C allele and CC homozygote of rs3806265 was more frequent in MG and RRMS patients than controls, whereas the T allele and TT homozygote were less frequent in RAS patients [40,41,42]. In the Chinese population, the T allele was associated with a higher risk of psoriasis and this locus might function as an enhancer in the immune-related system [43]. In our study, it was the opposite that C allele and CC homozygote were associated with a higher risk of IPA. This might be due to IPA occurring as a result of immune deficiency.
Previous research has found GG homozygote of rs212704 in NLRC4 was associated with A.fumigatus colonization in cystic fibrosis patients [24]. Our study showed that TT homozygote was associated with a decreased risk of aspergillosis, reflecting the role of NLRC4 in host defense against Aspergillus infection as well. Besides, C allele of rs212704 was associated with increased insulin and lower glucose levels while CC homozygote with a lower 2-h postprandial C-peptide level, suggesting that the polymorphism of NLRC4 may also relate to body metabolism [44,45].
The polymorphism of NLRC5 is also related to the susceptibility, severity, and prognosis of different kinds of diseases. In chronic periodontitis, the rs289723 in NLRC5 gene was associated with chronic slight and chronic localized periodontitis susceptibility and the AA genotype was correlated with increased risk of disease development [46]. It suggests that NLRC5 may play a promoting role in the insurgence of inflammation. So, it is reasonable that we found that the polymorphism of NLRC5 influenced the susceptibility to pulmonary aspergillosis, especially IPA. Some other studies reported that there was also an association between NLRC5 SNPs and the survival of colorectal and rectal cancer [47,48].
Our study demonstrated that the polymorphisms of NLRs (NLRP3, NLRC4, NLRC5) were associated with pulmonary aspergillosis risk, but there were still some limitations. First, it was a single-center study in southeastern China. So, it was hard to get a large case group, and our results might be influenced by geographic, ethnic, and genetic factors. Second, it was a retrospective study so we could not measure the NLRs expression or the levels of downstream inflammatory factors at the onset of aspergillosis. Likewise, we could only select healthy people as the control group instead of patients with the same underlying diseases and health condition. Finally, functional evaluations are needed to unveil the function of different SNPs of NLRs in the progression of aspergillosis.

5. Conclusions

Our results identified the association of NLRP3, NLRC4, and NLRC5 genetic variation with the susceptibility of pulmonary aspergillosis for the first time. These NLRs are probably involved in host immune defense against Aspergillus infection. Their function and mechanism in aspergillosis have not yet been thoroughly studied. Our results can provide a reference for studies on the role of NLRs in aspergillosis and other fungal infections.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm11071870/s1, Table S1: Genotype distribution of NLRs SNPs in pulmonary aspergillosis cases and controls; TableS2: Genotype distribution of NLRs SNPs in non-ABPA group, ABPA group and controls; Table S3: Genotype distribution of NLRs SNPs in IPA group, CPA group and controls.

Author Contributions

Conceptualization, L.L.; Data curation, J.Z. (Jinjin Zhong); Formal analysis, J.Z. (Jinjin Zhong); Funding acquisition, X.S.; Investigation, J.Z. (Jinjin Zhong), L.L., Y.L., Y.G., J.Z. (Jiangnan Zhao), B.C. and W.Z.; Methodology, J.Z. (Jinjin Zhong) and L.L.; Project administration, X.S.; Writing—original draft, J.Z. (Jinjin Zhong); Writing—review & editing, L.L. and X.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Project of Natural Science Foundation of China (82070011), and the Key Project of Jiangsu Commission of Health (K2019004).

Institutional Review Board Statement

The study was approved by the ethics committee of Jinling Hospital (2015NJKY-035-02).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study. Written informed consent has been obtained from the patient(s) to publish this paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Arastehfar, A.; Carvalho, A.; Houbraken, J.; Lombardi, L.; Garcia-Rubio, R.; Jenks, J.D.; Rivero-Menendez, O.; Aljohani, R.; Jacobsen, I.D.; Berman, J.; et al. Aspergillus fumigatus and aspergillosis: From basics to clinics. Stud. Mycol. 2021, 100, 100115. [Google Scholar] [CrossRef] [PubMed]
  2. Tekaia, F.; Latge, J.P. Aspergillus fumigatus: Saprophyte or pathogen? Curr. Opin. Microbiol. 2005, 8, 385–392. [Google Scholar] [CrossRef] [PubMed]
  3. Kosmidis, C.; Denning, D.W. The clinical spectrum of pulmonary aspergillosis. Thorax 2015, 70, 270–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Latge, J.P.; Chamilos, G. Aspergillus fumigatus and Aspergillosis in 2019. Clin. Microbiol. Rev. 2019, 33, e00140-18. [Google Scholar] [CrossRef] [PubMed]
  5. Plato, A.; Hardison, S.E.; Brown, G.D. Pattern recognition receptors in antifungal immunity. Semin. Immunopathol. 2015, 37, 97–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Cunha, C.; Carvalho, A. Genetic defects in fungal recognition and susceptibility to invasive pulmonary aspergillosis. Med. Mycol 2019, 57 (Suppl. 2), S211–S218. [Google Scholar] [CrossRef]
  7. Wojtowicz, A.; Bochud, P.Y. Host genetics of invasive Aspergillus and Candida infections. Semin. Immunopathol. 2015, 37, 173–186. [Google Scholar] [CrossRef] [PubMed]
  8. Bochud, P.Y.; Chien, J.W.; Marr, K.A.; Leisenring, W.M.; Upton, A.; Janer, M.; Rodrigues, S.D.; Li, S.; Hansen, J.A.; Zhao, L.P.; et al. Toll-like receptor 4 polymorphisms and aspergillosis in stem-cell transplantation. N. Engl. J. Med. 2008, 359, 1766–1777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Fisher, C.E.; Hohl, T.M.; Fan, W.; Storer, B.E.; Levine, D.M.; Zhao, L.P.; Martin, P.J.; Warren, E.H.; Boeckh, M.; Hansen, J.A. Validation of single nucleotide polymorphisms in invasive aspergillosis following hematopoietic cell transplantation. Blood 2017, 129, 2693–2701. [Google Scholar] [CrossRef] [PubMed]
  10. He, Q.; Li, H.; Rui, Y.; Liu, L.; He, B.; Shi, Y.; Su, X. Pentraxin 3 Gene Polymorphisms and Pulmonary Aspergillosis in Chronic Obstructive Pulmonary Disease Patients. Clin. Infect. Dis. 2018, 66, 261–267. [Google Scholar] [CrossRef] [PubMed]
  11. Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kale, S.D.; Ayubi, T.; Chung, D.; Tubau-Juni, N.; Leber, A.; Dang, H.X.; Karyala, S.; Hontecillas, R.; Lawrence, C.B.; Cramer, R.A.; et al. Modulation of Immune Signaling and Metabolism Highlights Host and Fungal Transcriptional Responses in Mouse Models of Invasive Pulmonary Aspergillosis. Sci. Rep. 2017, 7, 17096. [Google Scholar] [CrossRef] [PubMed]
  13. Mangan, M.S.J.; Olhava, E.J.; Roush, W.R.; Seidel, H.M.; Glick, G.D.; Latz, E. Targeting the NLRP3 inflammasome in inflammatory diseases. Nat. Rev. Drug Discov. 2018, 17, 588–606. [Google Scholar] [CrossRef] [PubMed]
  14. Swanson, K.V.; Deng, M.; Ting, J.P. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef]
  15. Briard, B.; Fontaine, T.; Samir, P.; Place, D.E.; Muszkieta, L.; Malireddi, R.K.S.; Karki, R.; Christgen, S.; Bomme, P.; Vogel, P.; et al. Galactosaminogalactan activates the inflammasome to provide host protection. Nature 2020, 588, 688–692. [Google Scholar] [CrossRef] [PubMed]
  16. Karki, R.; Man, S.M.; Malireddi, R.K.S.; Gurung, P.; Vogel, P.; Lamkanfi, M.; Kanneganti, T.D. Concerted activation of the AIM2 and NLRP3 inflammasomes orchestrates host protection against Aspergillus infection. Cell Host Microbe 2015, 17, 357–368. [Google Scholar] [CrossRef] [Green Version]
  17. Bauer, R.; Rauch, I. The NAIP/NLRC4 inflammasome in infection and pathology. Mol. Asp. Med. 2020, 76, 100863. [Google Scholar] [CrossRef]
  18. Meissner, T.B.; Li, A.; Biswas, A.; Lee, K.H.; Liu, Y.J.; Bayir, E.; Iliopoulos, D.; van den Elsen, P.J.; Kobayashi, K.S. NLR family member NLRC5 is a transcriptional regulator of MHC class I genes. Proc. Natl. Acad. Sci. USA 2010, 107, 13794–13799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Downs, I.; Vijayan, S.; Sidiq, T.; Kobayashi, K.S. CITA/NLRC5: A critical transcriptional regulator of MHC class I gene expression. Biofactors 2016, 42, 349–357. [Google Scholar] [CrossRef] [PubMed]
  20. Biswas, A.; Meissner, T.B.; Kawai, T.; Kobayashi, K.S. Cutting edge: Impaired MHC class I expression in mice deficient for Nlrc5/class I transactivator. J. Immunol. 2012, 189, 516–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Lupfer, C.R.; Stokes, K.L.; Kuriakose, T.; Kanneganti, T.D. Deficiency of the NOD-Like Receptor NLRC5 Results in Decreased CD8(+) T Cell Function and Impaired Viral Clearance. J. Virol. 2017, 91, e00377-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Sun, T.; Ferrero, R.L.; Girardin, S.E.; Gommerman, J.L.; Philpott, D.J. NLRC5 deficiency has a moderate impact on immunodominant CD8(+) T-cell responses during rotavirus infection of adult mice. Immunol. Cell Biol. 2019, 97, 552–562. [Google Scholar] [CrossRef] [PubMed]
  23. Gresnigt, M.S.; Cunha, C.; Jaeger, M.; Goncalves, S.M.; Malireddi, R.K.S.; Ammerdorffer, A.; Lubbers, R.; Oosting, M.; Rasid, O.; Jouvion, G.; et al. Genetic deficiency of NOD2 confers resistance to invasive aspergillosis. Nat. Commun. 2018, 9, 2636. [Google Scholar] [CrossRef]
  24. Iannitti, R.G.; Napolioni, V.; Oikonomou, V.; De Luca, A.; Galosi, C.; Pariano, M.; Massi-Benedetti, C.; Borghi, M.; Puccetti, M.; Lucidi, V.; et al. IL-1 receptor antagonist ameliorates inflammasome-dependent inflammation in murine and human cystic fibrosis. Nat. Commun. 2016, 7, 10791. [Google Scholar] [CrossRef] [PubMed]
  25. Patterson, T.F.; Thompson, G.R., 3rd; Denning, D.W.; Fishman, J.A.; Hadley, S.; Herbrecht, R.; Kontoyiannis, D.P.; Marr, K.A.; Morrison, V.A.; Nguyen, M.H.; et al. Practice Guidelines for the Diagnosis and Management of Aspergillosis: 2016 Update by the Infectious Diseases Society of America. Clin. Infect. Dis. 2016, 63, e1–e60. [Google Scholar] [CrossRef] [PubMed]
  26. Alexander, B.D.; Lamoth, F.; Heussel, C.P.; Prokop, C.S.; Desai, S.R.; Morrissey, C.O.; Baddley, J.W. Guidance on Imaging for Invasive Pulmonary Aspergillosis and Mucormycosis: From the Imaging Working Group for the Revision and Update of the Consensus Definitions of Fungal Disease from the EORTC/MSGERC. Clin. Infect. Dis. 2021, 72 (Suppl. 2), S79–S88. [Google Scholar] [CrossRef] [PubMed]
  27. Barrett, J.C.; Fry, B.; Maller, J.; Daly, M.J. Haploview: Analysis and visualization of LD and haplotype maps. Bioinformatics 2005, 21, 263–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Wigginton, J.E.; Cutler, D.J.; Abecasis, G.R. A note on exact tests of Hardy-Weinberg equilibrium. Am. J. Hum. Genet. 2005, 76, 887–893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Kesh, S.; Mensah, N.Y.; Peterlongo, P.; Jaffe, D.; Hsu, K.; Van Den Brink, M.; O’Reilly, R.; Pamer, E.; Satagopan, J.; Papanicolaou, G.A. TLR1 and TLR6 polymorphisms are associated with susceptibility to invasive aspergillosis after allogeneic stem cell transplantation. Ann. N. Y. Acad. Sci. 2005, 1062, 95–103. [Google Scholar] [CrossRef] [PubMed]
  30. Sainz, J.; Salas-Alvarado, I.; Lopez-Fernandez, E.; Olmedo, C.; Comino, A.; Garcia, F.; Blanco, A.; Gomez-Lopera, S.; Oyonarte, S.; Bueno, P.; et al. TNFR1 mRNA expression level and TNFR1 gene polymorphisms are predictive markers for susceptibility to develop invasive pulmonary aspergillosis. Int. J. Immunopathol. Pharmacol. 2010, 23, 423–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Grube, M.; Loeffler, J.; Mezger, M.; Kruger, B.; Echtenacher, B.; Hoffmann, P.; Edinger, M.; Einsele, H.; Andreesen, R.; Holler, E. TLR5 stop codon polymorphism is associated with invasive aspergillosis after allogeneic stem cell transplantation. Med. Mycol. 2013, 51, 818–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Sainz, J.; Lupianez, C.B.; Segura-Catena, J.; Vazquez, L.; Rios, R.; Oyonarte, S.; Hemminki, K.; Forsti, A.; Jurado, M. Dectin-1 and DC-SIGN polymorphisms associated with invasive pulmonary Aspergillosis infection. PLoS ONE 2012, 7, e32273. [Google Scholar] [CrossRef]
  33. Lupianez, C.B.; Canet, L.M.; Carvalho, A.; Alcazar-Fuoli, L.; Springer, J.; Lackner, M.; Segura-Catena, J.; Comino, A.; Olmedo, C.; Rios, R.; et al. Polymorphisms in Host Immunity-Modulating Genes and Risk of Invasive Aspergillosis: Results from the AspBIOmics Consortium. Infect. Immun. 2015, 84, 643–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Seo, K.W.; Kim, D.H.; Sohn, S.K.; Lee, N.Y.; Chang, H.H.; Kim, S.W.; Jeon, S.B.; Baek, J.H.; Kim, J.G.; Suh, J.S.; et al. Protective role of interleukin-10 promoter gene polymorphism in the pathogenesis of invasive pulmonary aspergillosis after allogeneic stem cell transplantation. Bone Marrow Transplant. 2005, 36, 1089–1095. [Google Scholar] [CrossRef] [PubMed]
  35. Lupianez, C.B.; Villaescusa, M.T.; Carvalho, A.; Springer, J.; Lackner, M.; Sanchez-Maldonado, J.M.; Canet, L.M.; Cunha, C.; Segura-Catena, J.; Alcazar-Fuoli, L.; et al. Common Genetic Polymorphisms within NFkappaB-Related Genes and the Risk of Developing Invasive Aspergillosis. Front. Microbiol. 2016, 7, 1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Zarrinfar, H.; Mirhendi, H.; Fata, A.; Khodadadi, H.; Kordbacheh, P. Detection of Aspergillus flavus and A. fumigatus in Bronchoalveolar Lavage Specimens of Hematopoietic Stem Cell Transplants and Hematological Malignancies Patients by Real-Time Polymerase Chain Reaction, Nested PCR and Mycological Assays. Jundishapur J. Microbiol. 2015, 8, e13744. [Google Scholar]
  37. Zarrinfar, H.; Makimura, K.; Satoh, K.; Khodadadi, H.; Mirhendi, H. Incidence of pulmonary aspergillosis and correlation of conventional diagnostic methods with nested PCR and real-time PCR assay using BAL fluid in intensive care unit patients. J. Clin. Lab. Anal. 2013, 27, 181–185. [Google Scholar] [CrossRef] [PubMed]
  38. Elinav, E.; Strowig, T.; Henao-Mejia, J.; Flavell, R.A. Regulation of the antimicrobial response by NLR proteins. Immunity 2011, 34, 665–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Schroder, K.; Tschopp, J. The inflammasomes. Cell 2010, 140, 821–832. [Google Scholar] [CrossRef] [Green Version]
  40. Imani, D.; Azimi, A.; Salehi, Z.; Rezaei, N.; Emamnejad, R.; Sadr, M.; Izad, M. Association of nod-like receptor protein-3 single nucleotide gene polymorphisms and expression with the susceptibility to relapsing-remitting multiple sclerosis. Int. J. Immunogenet. 2018, 45, 329–336. [Google Scholar] [CrossRef] [PubMed]
  41. Bidoki, A.Z.; Harsini, S.; Sadr, M.; Soltani, S.; Mohammadzadeh, M.; Najafi, S.; Rezaei, N. NLRP3 gene polymorphisms in Iranian patients with recurrent aphthous stomatitis. J. Oral Pathol. Med. 2016, 45, 136–140. [Google Scholar] [CrossRef] [PubMed]
  42. Agah, E.; Nafissi, S.; Saleh, F.; Sarraf, P.; Tafakhori, A.; Mousavi, S.V.; Saghazadeh, A.; Sadr, M.; Sinaei, F.; Mohebbi, B.; et al. Investigating the possible association between NLRP3 gene polymorphisms and myasthenia gravis. Muscle Nerve 2021, 63, 730–736. [Google Scholar] [CrossRef] [PubMed]
  43. Yu, P.; Hao, S.; Zheng, H.; Zhao, X.; Li, Y. Association of NLRP1 and NLRP3 Polymorphisms with Psoriasis Vulgaris Risk in the Chinese Han Population. BioMed Res. Int. 2018, 2018, 4714836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Xu, L.; Sun, X.; Xia, Y.; Luo, S.; Lin, J.; Xiao, Y.; Liu, Y.; Wang, Y.; Huang, G.; Li, X.; et al. Polymorphisms of the NLRC4 Gene are Associated with the Onset Age, Positive Rate of GADA and 2-h Postprandial C-Peptide in Patients with Type 1 Diabetes. Diabetes Metab. Syndr. Obes. 2020, 13, 811–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Gomes Torres, A.; Leite, N.; Tureck, L.V.; de Souza, R.L.R.; Titski, A.C.K.; Milano-Gai, G.E.; Lazarotto, L.; da Silva, L.R.; Furtado-Alle, L. Association between Toll-like receptors (TLR) and NOD-like receptor (NLR) polymorphisms and lipid and glucose metabolism. Gene 2019, 685, 211–221. [Google Scholar] [CrossRef]
  46. Zupin, L.; Navarra, C.O.; Robino, A.; Bevilacqua, L.; Di Lenarda, R.; Gasparini, P.; Crovella, S. NLRC5 polymorphism is associated with susceptibility to chronic periodontitis. Immunobiology 2017, 222, 704–708. [Google Scholar] [CrossRef] [PubMed]
  47. Catalano, C.; da Silva Filho, M.I.; Frank, C.; Jiraskova, K.; Vymetalkova, V.; Levy, M.; Liska, V.; Vycital, O.; Naccarati, A.; Vodickova, L.; et al. Investigation of single and synergic effects of NLRC5 and PD-L1 variants on the risk of colorectal cancer. PLoS ONE 2018, 13, e0192385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Catalano, C.; da Silva Filho, M.I.; Jiraskova, K.; Vymetalkova, V.; Levy, M.; Liska, V.; Vycital, O.; Naccarati, A.; Vodickova, L.; Hemminki, K.; et al. Short article: Influence of regulatory NLRC5 variants on colorectal cancer survival and 5-fluorouracil-based chemotherapy. Eur. J. Gastroenterol. Hepatol. 2018, 30, 838–842. [Google Scholar] [CrossRef]
Figure 1. A linkage disequilibrium (LD) plot of NLR SNPs for all aspergillosis patients and controls. The D’ value of each SNP pair is represented as a percentage and shown in the square (D’ ≥ 0.99 not shown). Red squares indicate strong pairwise linkage disequilibrium. (A) The two NLRP3 SNPs (rs3806265, rs7525979) constitute a haplotype block spanning 1 kb of the NLRP3 gene with an extremely high pair-wise D′ ≥ 0.99. (B) The three NLRC4 SNPs (rs385076, rs47933, rs7562653) constitute a haplotype block spanning 11 kb of the NLRC4 gene (C) The four NLRC5 SNPs (rs34531240, rs28438857, rs3995818, rs3995817) constitute a haplotype block spanning 7 kb of the NLRC5 gene with an extremely high pair-wise D′ ≥ 0.99.
Figure 1. A linkage disequilibrium (LD) plot of NLR SNPs for all aspergillosis patients and controls. The D’ value of each SNP pair is represented as a percentage and shown in the square (D’ ≥ 0.99 not shown). Red squares indicate strong pairwise linkage disequilibrium. (A) The two NLRP3 SNPs (rs3806265, rs7525979) constitute a haplotype block spanning 1 kb of the NLRP3 gene with an extremely high pair-wise D′ ≥ 0.99. (B) The three NLRC4 SNPs (rs385076, rs47933, rs7562653) constitute a haplotype block spanning 11 kb of the NLRC4 gene (C) The four NLRC5 SNPs (rs34531240, rs28438857, rs3995818, rs3995817) constitute a haplotype block spanning 7 kb of the NLRC5 gene with an extremely high pair-wise D′ ≥ 0.99.
Jcm 11 01870 g001
Table 1. Demographic and clinical characteristics of the study population.
Table 1. Demographic and clinical characteristics of the study population.
CharacteristicsCase Group (n = 73)Control Group (n = 103)p Value
Age58.48 ± 1.6355.25 ± 1.038n.s.
Gender (male/female)41/3264/39n.s.
Ethnicity n.s.
Han73 (100)103 (100)
Serum albumin, g/L32.23 ± 0.7314
Comorbidities
Hypertension20 (27.40)
Diabetes12 (16.44)
COPD11 (15.07)
Asthma5 (6.85)
Bronchiectasis23 (31.51)
Tuberculosis20 (27.40)
History of smoking21 (28.77)
Steroid treatment13 (17.81)
History of hepatitis infection3 (4.11)
Serum albumin < 30 g/L27 (36.99)
Classification
IPA30 (41.10)
CPA27 (36.99)
ABPA16 (21.92)
Table 2. Information about NLRP3, NLRC4, and NLRC5 SNPs in this study.
Table 2. Information about NLRP3, NLRC4, and NLRC5 SNPs in this study.
GeneSNP NumberChromosome PositionGene LocationMAFHWE in Control Group
NLRP3rs3806265247586336intron10.4720.0628
rs7525979247587408synon_exon30.1880.0381
rs35829419247588858nonsynon_exon30.01.0
rs107545582476120363′-UTR_exon100.4690.269
NLRC4rs12989936322685863′-flanking0.3471.0
rs21270432450348intron80.4660.7296
rs756265332478629intron20.4150.5904
rs47933332489158intron10.3441.0
rs38507632489851intron10.3920.9022
NLRC5rs12598522570223525′-flanking0.50.8838
rs3453124057060340synon_exon50.440.0882
rs2843885757060353nonsynon_exon50.440.0882
rs399581857068106nonsynon_exon120.4430.0882
rs399581757068107synon_exon120.440.0882
rs168457957071113synon_exon140.4230.1275
rs3751705571164583′-UTR_exon480.3270.5313
Table 3. Genotype distribution of NLRP3 rs3806265, NLRC4 rs212704, NLRC5 rs1684579, and rs12598522 in pulmonary aspergillosis cases and controls.
Table 3. Genotype distribution of NLRP3 rs3806265, NLRC4 rs212704, NLRC5 rs1684579, and rs12598522 in pulmonary aspergillosis cases and controls.
GeneSNPModelGenotypeCaseControlOR(95% CI)p Value
NLRP3rs3806265CodominantCC/CT/TT21/35/1714/61/28 0.0451 *
DominantCC+CT/TT56/1775/281.23 (0.6289 to 2.394)0.5593
RecessiveCC/CT+TT21/5214/892.567 (1.239 to 5.255)0.0130 *
AlleleC/T77/6989/1171.467 (0.9608 to 2.254)0.0774
NLRC4rs212704CodominantTT/CT/CC10/41/2227/49/27 0.1329
DominantTT+CT/CC51/2276/270.8236 (0.4285 to 1.578)0.5672
RecessiveTT/CT+CC10/6327/760.4468 (0.2071 to 0.959)0.0447 *
AlleleT/C61/85103/1030.7176 (0.4649 to 1.097)0.1278
NLRC5rs12598522CodominantTT/CT/CC23/37/1320/53/30 0.09069
DominantTT+CT/CC60/1373/301.897 (0.9209 to 3.82)0.0851
RecessiveTT/CT+CC23/5020/831.909 (0.957 to 3.897)0.0659
AlleleT/C83/6393/1131.601 (1.048 to 2.47)0.0305 *
rs1684579CodominantCC/CT/TT12/31/3017/60/26 0.0661
DominantCC+CT/TT43/3077/260.484 (0.2489 to 0.9213)0.0261 *
RecessiveCC/CT+TT12/6117/860.9952 (0.4295 to 2.301)0.9907
AlleleC/T55/9194/1120.7201 (0.4725 to 1.111)0.1364
*, a p value < 0.05 indicated a statistically significant difference.
Table 4. Genotype distribution of NLRP3 rs3806265, NLRC5 rs1684579 and rs12598522 in non-ABPA group and controls.
Table 4. Genotype distribution of NLRP3 rs3806265, NLRC5 rs1684579 and rs12598522 in non-ABPA group and controls.
GeneSNPModelNon-ABPA GroupControl GroupOR(95% CI)p Value
NLRP3rs3806265Codominant17/24/1614/61/28 0.0298 *
Dominant41/1675/280.9567 (0.4636 to 1.977)0.9044
Recessive17/4014/892.702 (1.172 to 5.924)0.0129 *
Allele58/5689/1171.362 (0.8541 to 2.179)0.1872
NLRC5rs12598522Codominant17/31/920/53/30 0.1077
Dominant48/973/302.192 (0.9867 to 5.247)0.0599
Recessive17/4020/831.764 (0.829 to 3.663)0.1349
Allele65/4993/1131.612 (1.012 to 2.529)0.0419 *
rs1684579Codominant9/21/2717/60/26 0.0125 *
Dominant30/2777/260.3752 (0.1889 to 0.7292)0.0044 **
Recessive9/4817/860.9485 (0.4001 to 2.237)0.9065
Allele39/7594/1120.6196 (0.381 to 1.006)0.0471 *
*, a p value < 0.05; **, a p value < 0.01.
Table 5. Genotype distribution of NLRP3 rs3806265 and NLRC5 rs34531240, rs28438857, rs3995818, rs3995817, rs1684579 in IPA group and controls.
Table 5. Genotype distribution of NLRP3 rs3806265 and NLRC5 rs34531240, rs28438857, rs3995818, rs3995817, rs1684579 in IPA group and controls.
GeneSNPModelIPA GroupControl GroupOR(95% CI)p Value
NLRP3rs3806265Codominant13/11/614/61/28 0.0017 **
Dominant24/675/281.493 (0.5538 to 3.979)0.4273
Recessive13/1714/894.861 (2.007 to 11.9)0.0004 ***
Allele37/2389/1172.115 (1.196 to 3.741)0.0117 *
NLRC5rs34531240Codominant4/13/1317/61/25 0.1251
Dominant17/1378/250.4191 (0.1796 to 1.026)0.0420 *
Recessive4/2617/860.7783 (0.2655 to 2.484)0.8928
Allele21/3995/1110.6291 (0.346 to 1.122)0.1265
rs28438857Codominant4/13/1317/61/25 0.1251
Dominant17/1378/250.4191 (0.1796 to 1.026)0.0420 *
Recessive4/2617/860.7783 (0.2655 to 2.484)0.8928
Allele21/3995/1110.6291 (0.346 to 1.122)0.1265
rs3995818Codominant4/13/1317/61/25 0.1251
Dominant17/1378/250.4191 (0.1796 to 1.026)0.0420 *
Recessive4/2617/860.7783 (0.2655 to 2.484)0.8928
Allele21/3995/1110.6291 (0.346 to 1.122)0.1265
rs3995817Codominant4/13/1317/61/25 0.1251
Dominant17/1378/250.4191 (0.1796 to 1.026)0.0420 *
Recessive4/2617/860.7783 (0.2655 to 2.484)0.8928
Allele21/3995/1110.6291 (0.346 to 1.122)0.1265
rs1684579Codominant5/9/1617/60/26 0.0092 **
Dominant14/1677/260.2955 (0.1234 to 0.6804)0.0036 **
Recessive5/2517/861.012 (0.3787 to 2.931)0.9833
Allele19/4194/1120.5522 (0.3026 to 1)0.0541
*, a p value < 0.05; **, a p value < 0.01; ***, a p value < 0.001.
Table 6. Haplotype analysis for NLRP3 gene polymorphisms in IPA group.
Table 6. Haplotype analysis for NLRP3 gene polymorphisms in IPA group.
Haplotypers34531240rs28438857TotalCaseControlp Value
H1TC0.5260.3830.5680.0117 *
H2CC0.2890.4170.2520.0136 *
H3CT0.1840.2000.1800.72
*, a p value < 0.05 indicated a statistically significant difference.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhong, J.; Liu, L.; Lu, Y.; Gu, Y.; Zhao, J.; Chen, B.; Zhou, W.; Su, X. NLRP3, NLRC4 and NLRC5 Gene Polymorphisms Associate with Susceptibility of Pulmonary Aspergillosis in Non-Neutropenic Patients. J. Clin. Med. 2022, 11, 1870. https://doi.org/10.3390/jcm11071870

AMA Style

Zhong J, Liu L, Lu Y, Gu Y, Zhao J, Chen B, Zhou W, Su X. NLRP3, NLRC4 and NLRC5 Gene Polymorphisms Associate with Susceptibility of Pulmonary Aspergillosis in Non-Neutropenic Patients. Journal of Clinical Medicine. 2022; 11(7):1870. https://doi.org/10.3390/jcm11071870

Chicago/Turabian Style

Zhong, Jinjin, Lulu Liu, Yajie Lu, Yu Gu, Jiangnan Zhao, Bilin Chen, Wei Zhou, and Xin Su. 2022. "NLRP3, NLRC4 and NLRC5 Gene Polymorphisms Associate with Susceptibility of Pulmonary Aspergillosis in Non-Neutropenic Patients" Journal of Clinical Medicine 11, no. 7: 1870. https://doi.org/10.3390/jcm11071870

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop