Skip to main content

Significance of methylation-related genes in diagnosis and subtype classification of renal interstitial fibrosis

Abstract

Background

RNA methylation modifications, such as N1-methyladenosine/N6-methyladenosine /N5-methylcytosine (m1A/m6A/m5C), are the most common RNA modifications and are crucial for a number of biological processes. Nonetheless, the role of RNA methylation modifications of m1A/m6A/m5C in the pathogenesis of renal interstitial fibrosis (RIF) remains incompletely understood.

Methods

Firstly, we downloaded 2 expression datasets from the GEO database, namely GSE22459 and GSE76882. In a differential analysis of these datasets between patients with and without RIF, we selected 33 methylation-related genes (MRGs). We then applied a PPI network, LASSO analysis, SVM-RFE algorithm, and RF algorithm to identify key MRGs.

Results

We eventually obtained five candidate MRGs (WTAP, ALKBH5, YTHDF2, RBMX, and ELAVL1) to forecast the risk of RIF. We created a nomogram model derived from five key MRGs, which revealed that the nomogram model may be advantageous to patients. Based on the selected five significant MRGs, patients with RIF were classified into two MRG patterns using consensus clustering, and the correlation between the five MRGs, the two MRG patterns, and the genetic pattern with immune cell infiltration was shown. Moreover, we conducted GO and KEGG analyses on 768 DEGs between MRG clusters A and B to look into their different involvement in RIF. To measure the MRG patterns, a PCA algorithm was developed to determine MRG scores for each sample. The MRG scores of the patients in cluster B were higher than those in cluster A.

Conclusions

Ultimately, we concluded that cluster A in the two MRG patterns identified on these five key m1A/m6A/m5C regulators may be associated with RIF.

Introduction

Low public awareness, high prevalence, high medical expenses, and a poor prognosis are all features of the significant public health threat known as chronic kidney disease (CKD), which affects people all over the world [1]. 697.5 million individuals worldwide, or 9.1% of the population, had CKD in 2017 [2]. Renal interstitial fibrosis (RIF) is a common pathological response to the widespread incidence of CKD. RIF is defined by the abnormal accumulation of extracellular matrix in the interstitial compartment situated between the tubules and peritubular capillaries, leading to tissue damage and functional impairment. Ultimately, this accumulation of matrix results in renal failure and eventual mortality [3, 4]. Unfortunately, there are no targeted, definitive, and effective treatments for RIF, so early recognition of patients at high risk for RIF is above all important for early prevention, delay, and reversal of the disease.

RNA modifications that occur in cells play an important role in regulating their stability, processing, transport, and gene expression and are the central switch in RNA metabolism [5]. RNA methylation, as the most typical modification in RNA modification, can be classified into various forms, including m1A, m6A and m5C, depending on the methylation site [6, 7]. Similar to histone modifications and DNA epigenetics, RNA methylation modifications can be installed, removed, and recognized by specific proteins called “writers,“ “erasers,“ and “readers”. The methylation modification known as m6A is formed by methylation of the sixth N atom of RNA adenine with the assistance of several m6A writers, including METTL3, METTL14, and WTAP [8,9,10]. Conversely, demethyltransferases (FTO and ALKBH5) that remove m6A, called m6A erasers, work with m6A writers to maintain a dynamic balance between mRNA methylation and unmethylation in cells [11]. YTHDF1/2/3 and YTHDC1/2 (YTH structural domain family proteins), as a category of RNA-binding proteins, recognize m6A modification sites that lead to changes in RNA metabolism [12, 13]. Under healthy circumstances, adenine is a positively electrostatically methylated nucleotide, and the m1A modification is a reversible methylation modification of the first nitrogen (N1) atom of adenine in RNA [14].

Similar to m6A, the m1A modification process is also regulated by m1A writers, erasers, and readers [11]. The m5C is a methylation alteration on the fifth carbon atom of cytosine that is extensively prevalent in a number of RNAs, including mRNA, tRNA, rRNA, and lncRNA. More than 10 RNA m5C methyltransferases have been discovered so far, including DNMT2, NSUN, and TRDMT [15]. The m5C site is primarily recognized by the reader proteins YBX1 and ALYREF [16, 17]. Erasers of m5A have not been identified, with only partial reports suggesting that the TET family has the potential to function as RNA demethylases [18]. In conclusion, the most common RNA methylation modifications have their own unique regulators and complex regulatory mechanisms, and they play a crucial role in living organisms. Abnormal RNA modification can promote the occurrence and development of various tumors by regulating cell growth, differentiation, migration, and drug resistance, such as gastric cancer, liver cancer, colorectal cancer, etc. [11].

The alteration of the apparent transcriptional profile of m6A in UUO mouse’s kidney signifies the crucial role played by m6A in the regulation of renal interstitial RIF [19]. In their study, Cui and colleagues utilized m6A methylation profiling to elucidate the role of m6A methylation in modulating oxidative stress and cytoplasmic metabolism during the development of liver fibrosis. Notably, their findings revealed distinct patterns of m6A methylation that were enriched in immune response and apoptosis, which were associated with fibrosis regression [20]. Moreover, Li et al. discovered that the activation of cardiac fibroblasts is closely associated with elevated expression of METTL3. Cui and colleagues utilized m6A methylation profiling to elucidate the role of m6A methylation in modulating oxidative stress and cytoplasmic metabolism during the development of liver fibrosis [21]. Notably, their findings revealed distinct patterns of m6A methylation that were enriched in immune response and apoptosis, which were associated with fibrosis regression [22].Unfortunately, the function of the m1A/m6A/m5C regulators in RIF is still unknown.

In the present study, we examined the roles of m1A/m6A/m5C regulators in the diagnosis and subtype classification of RIF using the GSE22459 and GSE76882 datasets in the GEO. We are based on five key m1A/m6A/m5C regulators (ALKBH5, ELAVL1, RBMX, WTAP, and YTHDF2). A genetic model for predicting RIF susceptibility was established, and patients were shown to derive good clinical benefit from this model. Furthermore, we constructed two distinct MRG models that closely correlate with the expression of genes implicated in RIF, suggesting that MRGs hold promise as diagnostic and subtype classification markers for RIF, with potential implications for the early intervention and treatment of this disease.

Materials and methods

Acquisition of data

The GSE22459 and GSE76882 datasets from the GEO database contain 99 healthy patients without RIF and 135 patients with RIF [23, 24]. In light of the existing literature, we have identified a total of 48 genes responsible for regulating m1A, m6A, and m5C modifications (Supplementary Table 1) [25,26,27]. We employed a two-fold approach to preprocessing our data. Firstly, we applied a log2 transformation to each dataset and normalized the resulting expression values separately. Secondly, we merged the two datasets and utilized the “ComBat” function, a popular batch correction algorithm, to eliminate the batch effect. This process ensured that any system-specific variations between the two datasets were effectively removed, resulting in a more robust and reliable dataset for downstream analysis [28]. To identify the MRGs associated with RIF, we conducted differential analysis between patients with and without RIF. Specifically, we utilized the R package “limma” to analyze the differentially expressed MRGs between the two groups, adopting the Wilcoxon test with a significance threshold of p value less than 0.05.

Identification of key MRGs

To model RIF, we applied the protein-protein interaction (PPI) network, least absolute shrinkage and selection operator (LASSO) analysis, support vector machine recursive feature elimination (SVM-RFE) algorithm, and random forest (RF) algorithm to identify key MRGs. To establish the PPI network, we initially imported 48 genes previously identified as m1A/m6A/m5C regulators into the STRING protein database. We subsequently applied a minimum interaction score threshold of 0.4, followed by utilization of cytoscape and the MCODE algorithm to identify the central network and hub genes [29, 30]. A higher interaction score indicated a more definite protein-protein relationship; an interaction score of 0.4 was the default parameter in the STRING database, and most previous research had been screened using this as the threshold [31, 32]. The LASSO analysis is a dimensionality reduction algorithm that exhibits superior performance compared to regression analysis [33]. We used a 10-fold cross-validation lasso analysis to screen for hub genes. The SVM-RFE algorithm is a machine learning technique that involves the use of SVMs for generating feature vectors and iteratively eliminating them to identify the optimal set of variables [34]. The RF algorithm is a component-based supervised learning method, often viewed as an extension of decision trees [35]. We used the “random forest” package in R software to build a RF model (ntree = 500) to select key regulator genes from differentially expressed MRGs. Finally, the hub MRGs were identified by taking the intersection of the four analysis methods.

Establishment of a nomogram model

We developed a nomogram model utilizing five candidate MRGs and utilized the “rms” package in R to predict the incidence of interstitial fibrosis in patients. Furthermore, we created calibration curves to examine the agreement between projected and actual results. Finally, to determine whether the model would benefit patients, we ran a decision curve analysis (DCA) and exhibited clinical effect curves [36, 37].

Identification of molecular subtype

To gain deeper insights into the molecular mechanisms of RIF, we employed the consensus clustering algorithm, a powerful tool that identifies subgroups of similar patterns within a given dataset. Utilizing the R package “Consensus Cluster Plus”, we conducted unsupervised clustering analysis to identify the distinct patterns of MRGs based on their expression profiles. Through the use of resampling techniques, we were able to validate the clusters generated and establish the robustness of our analyses [38]. The selection of the number of clusters involves consideration of three primary criteria: small intra-group differences, large inter-group differences, and sufficient sample sizes. We employed various methods, including consensus matrix plots, consensus cumulative distribution function (CDF) plots, relative changes in area under the CDF curve, and trace plots, to determine the optimal number of clusters. Subsequently, we generated histograms to visualize the expression differences in the five significant MRGs between the two MRG clusters.

Estimation of the genetic profile

In order to quantify the MRG patterns identified from our clustering analysis, we employed the principal component analysis (PCA) algorithm to calculate the MRG scores for each RIF sample. Specifically, we used the first and second principal components (PC1 and PC2, respectively) as representative scores for each sample. To compute an MRG score for each RIF patient, we used the following equation: MRG Score = Σ(PC1i + PC2i), where i represents the expression values of MRGs. This allowed us to generate a comprehensive score index that represented the MRG expression patterns for each RIF patient, providing a quantitative basis for subsequent analyses and interpretations.

Estimation of the immune cell infiltration

Single-sample gene set enrichment analysis (ssGSEA) has been widely used in the field of immunological research to quantify the relative abundance of specific immune cell populations present within complex biological samples. As such, ssGSEA has emerged as a powerful tool for evaluating the immune microenvironment in various disease states, including RIF. By analyzing gene expression data obtained from RIF patient samples, we were able to utilize ssGSEA to establish the abundance of immune cells present within each sample [39]. This information may contribute towards a better understanding of the role that immune cells play in driving RIF pathogenesis, and could potentially inform the development of novel immunotherapeutic approaches for the treatment of RIF.

Functional and pathway enrichment analysis

To further investigate the biological significance of our findings, we utilized a number of R packages, including “clusterProfiler”, “ComplexHeatmap”, “limma”, “colorspace”, “stringi”, and “ggplot2”, to conduct GO and KEGG enrichment analyses. These analyses allowed us to identify the functional and molecular pathways associated with the differentially expressed genes (DEGs) between the two MRG clusters (A and B) we identified (adjusted P value < 0.05) [40, 41]. By examining the enriched pathways and functions, we were able to derive a richer understanding of the underlying biological mechanisms and potential therapeutic targets relevant to RIF.

Results

Landscape of the 48 m1A/m6A/m5C regulators

The present study depicts its research methodology in Fig. 1, which showcases the research flow. Additionally, Table 1 offers an overview of the primary R packages utilized in this study and the functions they implement. To examine the differential expression levels of 48 MRGs within patients with and without RIF, we employed the “limma” package within R followed by the generation of histograms to display the results. We discovered 33 differentially expressed MRGs, in which METTL3, WTAP, RBMX, YTHDF2, DNMT1, and other regulatory factors were over-expressed in patients with RIF, whereas ALKBH5, ELAVL1, TRMT61B, and other regulatory factors were down-regulated (Fig. 2A and Supplementary Table 2).

Fig. 1
figure 1

The research flow

Table 1 An overview of the primary R packages utilized in this study and the functions they implement
Fig. 2
figure 2

Identification of five key MRGs. (A) Landscape of the 48 MRGs in RIF. (B) 16 MRGs obtained from the PPI network. (C) 17 MRGs were identified by the LASSO analysis. (D) 15 MRGs were identified by the RF model. (E) Thirteen MRGs were selected by the SVM-RFE algorithm. (F) Five key MRGs (ALKBH5, ELAVL1, RBMX, WTAP, and YTHDF2) were finally obtained after applying four algorithm

Identification of of key MRGs

To further screen for MRGs significantly associated with RIF, we used the PPI network, LASSO analysis, SVM-RFE algorithm, and RF algorithm to identify key regulatory genes. Using the String database, we investigated the interaction network of 33 differentially expressed MRGs and visualized them using the cytoscape software. We obtained 16 MRGs based on interaction scores greater than 0.4 and the MCODE algorithm (Fig. 2B). Using the LASSO algorithm, we identified 17 MRGs from the 33 differentially expressed MRGs (Fig. 2C). The 15 MRGs were identified by constructing the RF model (Fig. 2D). The tenfold cross-validation curve shows that the accuracy of SVM-RFE is highest only when we select the 13 MRGs (Fig. 2E). Finally, we overlapped the results of the above three different algorithms and PPI network analysis to obtain five MRGs (ALKBH5, ELAVL1, RBMX, WTAP, and YTHDF2) that were significantly associated with RIF (Fig. 2F).

Construction of the nomogram

Based on our previous selection of five candidate MRGs, we built a nomogram model to predict the incidence of RIF (Fig. 3A). The calibration curve demonstrates that the nomogram model’s predictiveness is correct (Fig. 3B). Nomogram model-based decisions hold potential advantages for RIF patients, as evidenced by the DCA curve displaying the red line remaining consistently above the gray and black lines within the range of 0 to 1 (Fig. 3C). Furthermore, the clinical impact curve shows the nomogram model to possess robust predictive power (Fig. 3D).

Fig. 3
figure 3

(A) The construction of the nomogram model according to the five candidate MRGs. (B) The predictive ability of the nomogram model as exposed by the calibration curve. (C) Decisions based on the nomogram model may benefit patients with RIF. (D) The clinical impact of the nomogram model as assessed by the clinical impact curve

Identification of two different MRG patterns

Based on the five significant MRGs, two MRG patterns (MRG clusters A and B) were identified by using the “ConsensusClusterPlus” package in R software and the consensus clustering method. (Fig. 4A and Supplementary Fig. 1). We then plotted histograms to observe the differences in the expression levels of the 5 MRGs between these two MRG clusters (Fig. 4B). RBMX, WTAP, and YTHDF2 were expressed at higher levels in MRGcluster A than in MRGcluster B, while the opposite was true for ALKBH5. ELAVL1 expression did not differ significantly between MRG clusters A and B. PCA shows that the two MRG models can be fully distinguished based on these five important MRGs (Fig. 4C). We next calculated the immune cell abundance in the RIF samples using ssGSEA, and we produced histograms to demonstrate the various immune cell infiltrations between the two MRG clusters (Fig. 4D). In MRG cluster A, we discovered that activated B cells, activated CD4 T cells, activated CD8 T cells, activated dendritic T cells, eosinophils, gamma delta T cells, immature B cells, MDSCs, macrophages, mast cells, natural killer T cells, plasmacytoid dendritic cells, regulatory T cells, T follicular helper cells, type 1 T helper cells, and type 2 T helper cells were significantly expressed (p < 0.05). In addition, we evaluated the correlation between the five MRGs and immune cells separately and plotted the histogram (Fig. 4E). We found that WTAP was positively correlated with many immune cells, except CD56 bright natural killer cells, CD56 dim natural killer cells, and neutrophils. And ALKBH5 was negatively correlated with many immune cells, except CD56 dim natural killer cells, and immature dendritic cells. We classified the five key MRGs into high and low expression groups according to their median gene expression levels.

Fig. 4
figure 4

Consensus clustering of five significant MRGs. (A) Consensus matrices of five significant MRGs for k = 2. (B) A differential expression histogram of five noteworthy MRGs in MRG clusters A and B. (C) PCA for the expression profiles of five significant MRGs. (D) Differences in cell infiltration between MRG clusters A and B. (E) Connection between infiltrating immune cells and five significant MRGs

Thereafter, we further evaluated their correlation with immune cell infiltration (Fig. 5).

Fig. 5
figure 5

Correlation of the expression levels of five key MRGs with immune cell infiltration

Function and pathway enrichment

A total of 768 DEGs were selected in two MRG patterns with thresholds of | logFC | greater than or equal to 0.585 and an adjusted p value greater than 0.05. We used GO and KEGG enrichment analyses to further investigate the probable roles and molecular pathways of these DEGs in RIF. Biological process (BP) terminology is associated with the small molecule catabolic process (GO: 0044282) and the organic acid catabolic process (GO: 0016054); cellular component (CC) terminology is associated with apical plasma membrane (GO: 0016324) and apical part of cell (GO: 0045177); molecular function (MF) terms are related to secondary active transmembrane transporter activity (GO: 0015291) and oxidoreductase activity, acting on CH-OH group of donors (GO: 0016614)(Fig. 6A and Supplementary Table 3). KEGG enrichment analysis revealed that DEGs were highly enriched in the leishmaniasis (hsa05140) and chemokine signaling (hsa04062) pathways (Fig. 6B and Supplementary Table 4).

Fig. 6
figure 6

Consensus clustering of DEGs. (A and B) The GO and KEGG analyses for DEGs. (C) Consensus matrices of DEGs for k = 2. (D) A histogram of differential expression of the five key MRGs in gene clusters A and B. (E) Differences in immune cell infiltration between gene clusters A and B

Identification of two distinct genetic patterns and immune cell infiltration

To further verify the MRG pattern, patients with RIF were divided into different genomic subtypes based on 768 DEGs using a consistent clustering approach. We got two different gene patterns, gene cluster A and gene cluster B, which corresponded to the grouping of MRG patterns (Fig. 6C), while PCA revealed that the two gene patterns were fully distinct (Fig. 6E). Figure 6D F showed that the differential expression levels of the five key MRGs between gene cluster A and gene cluster B and the immune cell infiltration between the two gene patterns are similar to those in the MRG clusters. This again validates the correctness of the consensus clustering approach to grouping. To compute the MRG cluster, we used a PCA algorithm to compare the MRG scores between two different MRG clusters or gene clusters. The results showed that MRG cluster B, or gene cluster B, had a significantly higher MRG score than MRG cluster A, or gene cluster A (Fig. 7A). The relationship between MRG pattern, gene pattern, and MRG score is visualized in the Sankey plot (Fig. 7B).

Fig. 7
figure 7

Role of MRG patterns in the identification of RIF. (A) There were differences in MRG scores between MRG gene clusters A and B and between gene clusters A and B. (B) A Sankey diagram showing the relationship between MRG patterns, gene patterns, and MRG scores. (C) Differences in expression levels of RTEL1, MET, HNF1B, PAX2, TMEM67, CEP290, AGT, COPA, CLCN5, and AGTR1 between MRG clusters A and B and between gene clusters A and B

The role of MRG and gene patterns in RIF

To further investigate the connection between the MRG cluster and RIF, we explored the relationship between different clusters and the expression levels of genes closely related to the development of RIF, including RTEL1, MET, HNF1B, PAX2, TMEM67, CEP290, AGT, COPA, CLCN5, and AGTR1. The results showed that the expression levels of MET, TMEM67, CEP290, and COPA were higher in gene cluster A or MRG cluster A, while the rest were reversed, suggesting that gene cluster A or MRG cluster A is highly associated with RIF characteristics (Fig. 7 C and D).

Discussion

RIF is a common pathological manifestation of CKD progressing to end-stage renal disease. Its pathogenesis is mainly related to renal inflammatory injury, oxidative stress, and apoptosis, but the most important pathogenesis is the imbalance of extracellular matrix synthesis and/or degradation, and excessive deposition in normal interstitium and tubules [1, 42, 43]. RNA methylation has emerged as a crucial molecular mechanism involved in diverse biological pathways, including regulation of stem cell homeostasis, cell differentiation, DNA damage response, and gene expression. Moreover, dysregulation of RNA methylation has been implicated in the pathogenesis of multiple diseases, such as liver fibrosis, colorectal cancer, gastric cancer, bladder cancer, and several others [44,45,46]. However, no study has yet identified the role and mechanism of m1A, m6A and m5C modifications in RIF.

We first identified 33m1A/m6A/m5C regulators by differential expression analysis between patients with and without RIF. Then we finally obtained 5 key MRGs (ALKBH5, ELAVL1, RBMX, WTAP, and YTHDF2) by applying RF, LASSO, SVM, and PPI networks. Subsequently, a nomogram model was constructed utilizing 5 MRGs, enabling the prediction of RIF onset. The DCA curves suggest that decision-making based on the nomogram model could prove advantageous for patients experiencing RIF. Furthermore, two distinct MRG patterns were identified based on the aforementioned 5 MRGs. The correlation between immune cell infiltration, and these two patterns, and the five MRGs was subsequently explored. In between the two MRG patterns, we screened an additional 768 DEGs and distinguished two different gene patterns using consensus clustering, and interestingly, this gene pattern was grouped in line with the MRG pattern.

WTAP is a widespread nuclear protein that localizes throughout the nucleoplasm as well as in patches and binds specifically to the Wilm’s tumor 1 protein [47]. As an essential component of the methyltransferase complex, it can interact directly with METTL3 to recruit METTL3-METTL14 heterodimeric complexes to nuclear speckles for m6A modification [8]. Moreover, WTAP is involved in selective splicing of mRNA and cell cycle regulation [48]. Wei et al. found that AcSDKP could reduce the stability of Ptch1 mRNA by downregulating WTAP expression and ultimately exert anti-fibrotic effects [49]. A gene on the X chromosome codes for RBMX, a nuclear RNA-binding protein with a length of 43 kDa. It supports genomic integrity, transcription regulation, and splicing at the molecular level and is directly linked to healthy development, cancer, and viral infections [50]. Renieri et al. used genome sequencing to find mutations truncating the RBMX gene in lung cancer patients [51]. According to Martinez-Arribas et al., pro-apoptotic Bax gene expression and RBMX expression were associated with breast cancer [52]. ALKBH5 belongs to the AlkB subfamily of the 2OG dioxygenase superfamily, also known as erasers, and contains a highly conserved DSBH fold (also known as the jelly-roll motif) in its structure, which maintains the intracellular homeostasis of m6A modifications with the assistance of Fe2 + and the cofactor 2OG in conjunction with the methyltransferase complex [53]. ALKBH5 can affect the development of human malignant diseases by regulating biological processes [54, 55]. ALKBH5 can contribute to the progression of lung fibrosis by directly or indirectly regulating FOXM1 [56]. Inhibition of ALKBH5 in hepatic stellate cells attenuates radiation-induced liver fibrosis [57].

HuR, the gene product of the ELAVL1 gene, is the only member of the entire ELAVL family that is commonly expressed as RBPs in all human tissues [58]. HuR contains three RRMs, which are translocated from the nucleus to the cytoplasm when exposed to intrinsic and/or extrinsic stress and bind to A/U-rich elements in the 3’ untranslated region of mRNA to regulate mRNA splicing, transport, and stability [59, 60]. A prior investigation on liver fibrosis revealed that HuR binds to the 3’UTR of S1PR3 mRNA, thereby enhancing its stability through competitive inhibition of miR-30e. This resultant regulation of signaling pathways mediated via S1P-S1PR3 subsequently impacts the migration and differentiation of BMSCs into myofibroblasts [61]. Sorafenib attenuates hepatic fibrosis in mice by inducing hepatic stellate cell ferroptosis [62]. IAPF overexpression can block autophagy in lung fibrosis, and the action is dependent on ELAVL1 [63]. This all suggests a potential role for ELAVL1 and HuR in the development of liver fibrosis. As the most efficient m6A “reader”, YTHDF2 can target m6A-containing RNAs through its C-terminal YTH structural domain to regulate RNA processing, stabilization, and translation [64, 65]. YTHDF2 functions in various biological processes, including cancer development, inflammatory responses, regulation of hematopoietic stem cell self-renewal and differentiation, and initiation of pluripotent stem cell generation [64].In conclusion, the above five m1A/m6A/m5C regulators had direct or indirect relationships with fibrosis.

The pathogenesis of interstitial fibrosis is strongly connected to abnormalities in the immune system, particularly those related to the regulatory mechanisms of mast cells. A significant aspect of this process involves the synthesis, storage, and release of reactive renin by mast cells, which cleaves angiotensinogen and contributes to the production of ANG II, a key pro-fibrotic factor associated with renal fibrosis. Recent research conducted by Veerappan et al. demonstrated that treatment with sodium cromoglycate, a mast cell stabilizer, resulted in a reduction of collagen deposition and tubulointerstitial fibrosis in obstructed kidneys after a 14-day period [66]. This finding highlights the critical role of mast cells in the development of interstitial fibrosis. According to a study by Hirooka et al., Foxp3+-Treg has a protective function in the pathophysiology of RIF by activating the L-18R signaling pathway [67]. Wang et al. found by bone marrow transplantation experiments that bone marrow-derived macrophages, especially M2-type macrophages, could transdifferentiate into renal myofibroblasts and promote RIF [68]. TGF-β1 is an essential mediator in the pathogenesis of RIF, with anti-inflammatory and pro-fibrotic effects [69]. In fact, TGF-β1 is associated with a variety of immune cells, including B cells, CD8 + T cells, regulatory CD4 + T cells, and dendritic cells. For example, TGF-β1 can induce cell cycle arrest in B cells by inhibiting PI3K/Akt signaling, especially in the G0/G1 phase [70]. In addition, TGF-β1 can also indirectly impede B cell proliferation and activation by interacting with regulatory T cells [71]. In conclusion, given the importance of TGF-f1 in the immune cascade response, we know that the pathogenesis of RIF is associated with multiple immune cell infiltrations.

In our study, two MRG patterns were recognized by using a consensus clustering approach on five selected key MRGs. The high level of multiple immune cell infiltration in MRG cluster A suggests that MRG cluster A may be associated with RIF. The accuracy of the aforementioned results was then verified using DEGs connected to the gene pattern. By utilizing the PCA algorithm to determine the MRG score for each sample, we finally quantified the MRG pattern. Finally, we discovered that MRG cluster or gene cluster A had lower MRG scores. However, there are still some shortcomings in the study. First, since the datasets used do not have clinical data, including age, gender, glomerular filtration rate, etc., there is no way to further investigate the relationship between this clinical information and the different subtypes. Second, the sample size of this study is small, which may lead to some bias. Third, the results of the study still need to be validated by clinical cohort studies and in vitro and in vivo experiments. Therefore, we will collect more clinical samples, record more clinical information, and conduct in vitro and in vivo experiments and clinical cohort studies to further test our results.

Conclusion

To sum up, our investigation successfully identified five significant MRGs and developed a nomogram model to predict the incidence of RIF in patients. Additionally, two distinct MRG patterns were identified based on the aforementioned five MRGs, with MRG cluster A potentially linked to RIF. Collectively, our findings suggest that MRGs could serve as a viable diagnostic tool for identifying RIF subtypes, thus contributing to its prevention, delay, and treatment.

Data availability

The raw data used and analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Hockham C, Bao L, Tiku A, Badve SV, Bello AK, Jardine MJ, Jha V, Toyama T, Woodward M, Jun M. Sex differences in chronic kidney disease prevalence in Asia: a systematic review and meta-analysis. Clin kidney J. 2022;15(6):1144–51.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Liu XY, Zhang XB, Zhao YF, Qu K, Yu XY. Research Progress of Chinese Herbal Medicine intervention in renal interstitial fibrosis. Front Pharmacol. 2022;13:900491.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Herrera J, Henke CA, Bitterman PB. Extracellular matrix as a driver of progressive fibrosis. J Clin Investig. 2018;128(1):45–53.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Feng YL, Wang WB, Ning Y, Chen H, Liu P. Small molecules against the origin and activation of myofibroblast for renal interstitial fibrosis therapy. Volume 139. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie; 2021. p. 111386.

  5. Kadumuri RV, Janga SC. Epitranscriptomic Code and its alterations in Human Disease. Trends Mol Med. 2018;24(10):886–903.

    Article  CAS  PubMed  Google Scholar 

  6. Roundtree IA, Evans ME, Pan T, He C. Dynamic RNA modifications in Gene expression regulation. Cell. 2017;169(7):1187–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Xie S, Chen W, Chen K, Chang Y, Yang F, Lin A, Shu Q, Zhou T, Yan X. Emerging roles of RNA methylation in gastrointestinal cancers. Cancer Cell Int. 2020;20(1):585.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ping XL, Sun BF, Wang L, Xiao W, Yang X, Wang WJ, Adhikari S, Shi Y, Lv Y, Chen YS, et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014;24(2):177–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014;10(2):93–5.

    Article  CAS  PubMed  Google Scholar 

  10. Jurga S, Barciszewski J, Epitranscriptomics. RNA Technologies. 2021; 12.

  11. Zhang X, Su H, Chen H, Li Q, Liu X, Zhang L, Wu W, Chan M, Chen H. RNA modifications in gastrointestinal Cancer: current status and future perspectives. Biomedicines. 2022; 10(8).

  12. Shi HL, Wang X, Lu ZK, Zhao B, Ma HH, Hsu PJ, Liu C, He C. YTHDF3 facilitates translation and decay of N-6-methyladenosine-modified RNA. Cell Res. 2017;27(3):315–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wang X, Zhao BS, Roundtree IA, Lu ZK, Han DL, Ma HH, Weng XC, Chen K, Shi HL, He C. N-6-methyladenosine modulates Messenger RNA translation efficiency. Cell. 2015;161(6):1388–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Zhang C, Jia GF. Reversible RNA modification N-1 - methyladenosine (m(1)A) in mRNA and tRNA. Genom Proteom Bioinf. 2018;16(3):155–61.

    Article  CAS  Google Scholar 

  15. Chen YS, Yang WL, Zhao YL, Yang YG. Dynamic transcriptomic m(5)C and its regulatory role in RNA processing. Wires Rna. 2021; 12(4).

  16. Yang Y, Wang L, Han X, Yang WL, Zhang MM, Ma HL, Sun BF, Li A, Xia J, Chen J, et al. RNA 5-Methylcytosine facilitates the maternal-to-zygotic transition by preventing maternal mRNA decay. Mol Cell. 2019;75(6):1188.

    Article  CAS  PubMed  Google Scholar 

  17. Zou F, Tu RJ, Duan B, Yang ZL, Ping ZH, Song XQ, Chen SY, Price A, Li H, Scott A, et al. Drosophila YBX1 homolog YPS promotes ovarian germ line stem cell development by preferentially recognizing 5-methylcytosine RNAs. P Natl Acad Sci Usa. 2020;117(7):3603–9.

    Article  CAS  Google Scholar 

  18. Wang S, Li H, Lian Z, Deng S. The role of RNA modification in HIV-1 infection. Int J Mol Sci. 2022; 23(14).

  19. Li X, Fan X, Yin X, Liu H, Yang Y. Alteration of N-methyladenosine epitranscriptome profile in unilateral ureteral obstructive nephropathy. Epigenomics-Uk. 2020;12(14):1157–73.

    Article  CAS  Google Scholar 

  20. Cui Z, Huang N, Liu L, Li X, Li G, Chen Y, Wu Q, Zhang J, Long S, Wang M, et al. Dynamic analysis of m6A methylation spectroscopy during progression and reversal of hepatic fibrosis. Epigenomics-Uk. 2020;12(19):1707–23.

    Article  CAS  Google Scholar 

  21. Li T, Zhuang Y, Yang W, Xie Y, Shang W, Su S, Dong X, Wu J, Jiang W, Zhou Y, et al. Silencing of METTL3 attenuates cardiac fibrosis induced by myocardial infarction via inhibiting the activation of cardiac fibroblasts. Faseb J. 2021;35(2):e21162.

    Article  CAS  PubMed  Google Scholar 

  22. Zhang JX, Huang PJ, Wang DP, Yang WY, Lu J, Zhu Y, Meng XX, Wu X, Lin QH, Lv H, et al. M(6)a modification regulates lung fibroblast-to-myofibroblast transition through modulating KCNH6 mRNA translation. Mol Ther. 2021;29(12):3436–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Modena BD, Kurian SM, Gaber LW, Waalen J, Su AI, Gelbart T, Mondala TS, Head SR, Papp S, Heilman R, et al. Gene expression in biopsies of Acute rejection and interstitial Fibrosis/Tubular atrophy reveals highly Shared Mechanisms that Correlate with worse long-term outcomes. Am J Transplant. 2016;16(7):1982–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Park WD, Griffin MD, Cornell LD, Cosio FG, Stegall MD. Fibrosis with inflammation at one year predicts Transplant Functional decline. J Am Soc Nephrol. 2010;21(11):1987–97.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Li D, Li K, Zhang W, Yang KW, Mu DA, Jiang GJ, Shi RS, Ke D. The m6A/m5C/m1A regulated gene signature predicts the prognosis and correlates with the Immune Status of Hepatocellular Carcinoma. Front Immunol. 2022; 13.

  26. Shao DQ, Li Y, Wu JY, Zhang BB, Xie S, Zheng XL, Jiang ZQ. An m6A/m5C/m1A/m7G-Related long non-coding RNA signature to Predict Prognosis and Immune features of Glioma. Front Genet. 2022; 13.

  27. Rong DW, Sun GS, Wu F, Cheng Y, Sun GQ, Jiang W, Li X, Zhong Y, Wu LL, Zhang CY, et al. Epigenetics: roles and therapeutic implications of non-coding RNA modifications in human cancers. Mol Ther-Nucl Acids. 2021;25:67–82.

    Article  CAS  Google Scholar 

  28. Leek JT, Johnson WE, Parker HS, Jaffe AE, Storey JD. The sva package for removing batch effects and other unwanted variation in high-throughput experiments. Bioinformatics. 2012;28(6):882–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bader GD, Hogue CWV. An automated method for finding molecular complexes in large protein interaction networks. BMC Bioinformatics. 2003;4:2.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Szklarczyk D, Gable AL, Nastou KC, Lyon D, Kirsch R, Pyysalo S, Doncheva NT, Legeay M, Fang T, Bork P, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49:D605–12.

    Article  CAS  PubMed  Google Scholar 

  32. Mao J, Wang G, Yang L, Tan L, Tian C, Tang L, Fang L, Mu Z, Zhu Z, Li Y. Combined Network Pharmacology and Molecular Docking to verify the treatment of type 2 diabetes with Pueraria Lobata Radix and Salviae Miltiorrhizae Radix. Comput Math Methods Med. 2023;2023:9150324.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Tibshirani R. Regression shrinkage and selection via the lasso: a retrospective. J R Stat Soc B. 2011;73:273–82.

    Article  Google Scholar 

  34. Sanz H, Valim C, Vegas E, Oller JM, Reverter F. SVM-RFE: selection and visualization of the most relevant features through non-linear kernels. BMC Bioinformatics. 2018;19(1):432.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Mantas CJ, Castellano JG, Moral-Garcia S, Abellan J. A comparison of random forest based algorithms: random credal random forest versus oblique random forest. Soft Comput. 2019;23(21):10739–54.

    Article  Google Scholar 

  36. Iasonos A, Schrag D, Raj GV, Panageas KS. How to build and interpret a nomogram for cancer prognosis. J Clin oncology: official J Am Soc Clin Oncol. 2008;26(8):1364–70.

    Article  Google Scholar 

  37. Dai B, Sun F, Cai X, Li C, Liu H, Shang Y. Significance of RNA N6-Methyladenosine regulators in the diagnosis and subtype classification of Childhood Asthma using the gene expression Omnibus Database. Front Genet. 2021;12:634162.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wilkerson MD, Hayes DN. ConsensusClusterPlus: a class discovery tool with confidence assessments and item tracking. Bioinf (Oxford England). 2010;26(12):1572–3.

    CAS  Google Scholar 

  39. Zhang N, Zhao YD, Wang XM. CXCL10 an important chemokine associated with cytokine storm in COVID-19 infected patients. Eur Rev Med Pharmaco. 2020;24(13):7497–505.

    CAS  Google Scholar 

  40. Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innov (Camb). 2021;2(3):100141.

    CAS  Google Scholar 

  41. Gu Z, Eils R, Schlesner M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32(18):2847–9.

    Article  CAS  PubMed  Google Scholar 

  42. Jiang YF, Zhu YH, Zhen TM, Li J, Xing KC, He LQ, Zhu SB. Transcriptomic analysis of the mechanisms of alleviating renal interstitial fibrosis using the traditional chinese medicine Kangxianling in a rat model. Sci Rep-Uk. 2020; 10(1).

  43. Wang H, Jiang Q, Zhang L. Baicalin protects against renal interstitial fibrosis in mice by inhibiting the TGF-β/Smad signalling pathway. Pharm Biol. 2022;60(1):1407–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Pinello N, Sun S, Wong JJ. Aberrant expression of enzymes regulating mA mRNA methylation: implication in cancer. Cancer Biol Med. 2018;15(4):323–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhong H, Liu S, Cao F, Zhao Y, Zhou J, Tang F, Peng Z, Li Y, Xu S, Wang C, et al. Dissecting Tumor Antigens and Immune Subtypes of Glioma to develop mRNA vaccine. Front Immunol. 2021;12:709986.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Yin J, He X, Qin F, Zheng S, Huang Y, Hu L, Chen Y, Zhong L, Hu W, Li S. M(6)A-related lncRNA signature for predicting prognosis and immune response in head and neck squamous cell carcinoma. Am J Transl Res. 2022;14(11):7653–69.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Little NA, Hastie ND, Davies RC. Identification of WTAP, a novel Wilms’ tumour 1-associating protein. Hum Mol Genet. 2000;9(15):2231–9.

    Article  CAS  PubMed  Google Scholar 

  48. Horiuchi K, Kawamura T, Iwanari H, Ohashi R, Naito M, Kodama T, Hamakubo T. Identification of Wilms’ tumor 1-associating protein complex and its role in alternative splicing and the cell cycle. J Biol Chem. 2013;288(46):33292–302.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wei A, Zhao F, Hao A, Liu B, Liu Z. N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) mitigates the liver fibrosis via WTAP/mA/Ptch1 axis through hedgehog pathway. Gene. 2022;813:146125.

    Article  CAS  PubMed  Google Scholar 

  50. Elliott DJ, Dalgliesh C, Hysenaj G, Ehrmann I. RBMX family proteins connect the fields of nuclear RNA processing, disease and sex chromosome biology. Int J Biochem Cell Biol. 2019;108:1–6.

    Article  CAS  PubMed  Google Scholar 

  51. Renieri A, Mencarelli MA, Cetta F, Baldassarri M, Mari F, Furini S, Piu P, Ariani F, Dragani TA, Frullanti E. Oligogenic germline mutations identified in early non-smokers lung adenocarcinoma patients. Lung cancer (Amsterdam Netherlands). 2014;85(2):168–74.

    Article  PubMed  Google Scholar 

  52. Martínez-Arribas F, Agudo D, Pollán M, Gómez-Esquer F, Díaz-Gil G, Lucas R, Schneider J. Positive correlation between the expression of X-chromosome RBM genes (RBMX, RBM3, RBM10) and the proapoptotic bax gene in human breast cancer. J Cell Biochem. 2006;97(6):1275–82.

    Article  PubMed  Google Scholar 

  53. You Y, Fu Y, Huang M, Shen D, Zhao B, Liu H, Zheng Y, Huang L. Recent advances of m6A demethylases inhibitors and their Biological Functions in Human Diseases. Int J Mol Sci. 2022; 23(10).

  54. Li N, Kang Y, Wang L, Huff S, Tang R, Hui H, Agrawal K, Gonzalez GM, Wang Y, Patel SP, et al. ALKBH5 regulates anti-PD-1 therapy response by modulating lactate and suppressive immune cell accumulation in tumor microenvironment. P Natl Acad Sci Usa. 2020;117(33):20159–70.

    Article  CAS  Google Scholar 

  55. Shen C, Sheng Y, Zhu AC, Robinson S, Jiang X, Dong L, Chen H, Su R, Yin Z, Li W, et al. RNA demethylase ALKBH5 selectively promotes Tumorigenesis and Cancer Stem Cell Self-Renewal in Acute myeloid leukemia. Cell Stem Cell. 2020;27(1):64–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sun W, Li Y, Ma D, Liu Y, Xu Q, Cheng D, Li G, Ni C. ALKBH5 promotes lung fibroblast activation and silica-induced pulmonary fibrosis through miR-320a-3p and FOXM1. Cell Mol Biol Lett. 2022;27(1):26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Chen Y, Zhou P, Deng Y, Cai X, Sun M, Sun Y, Wu D. ALKBH5-mediated m a demethylation of TIRAP mRNA promotes radiation-induced liver fibrosis and decreases radiosensitivity of hepatocellular carcinoma. Clin translational Med. 2023;13(2):e1198.

    Article  CAS  Google Scholar 

  58. Hinman MN, Lou H. Diverse molecular functions of Hu proteins. Cell Mol Life Sci. 2008;65(20):3168–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Diaz-Quintana A, Garcia-Maurino SM, Diaz-Moreno I. Dimerization model of the C-terminal RNA Recognition Motif of HuR. Febs Lett. 2015;589(10):1059–66.

    Article  CAS  PubMed  Google Scholar 

  60. Schultz CW, Preet R, Dhir T, Dixon DA, Brody JR. Understanding and targeting the disease-related RNA binding protein human antigen R (HuR). Wiley interdisciplinary reviews. RNA. 2020;11(3):e1581.

    CAS  PubMed  Google Scholar 

  61. Chang N, Ge J, Xiu L, Zhao Z, Duan X, Tian L, Xie J, Yang L, Li L. HuR mediates motility of human bone marrow-derived mesenchymal stem cells triggered by sphingosine 1-phosphate in liver fibrosis. J Mol Med. 2017;95(1):69–82.

    Article  CAS  PubMed  Google Scholar 

  62. Zhang Z, Yao Z, Wang L, Ding H, Shao J, Chen A, Zhang F, Zheng S. Activation of ferritinophagy is required for the RNA-binding protein ELAVL1/HuR to regulate ferroptosis in hepatic stellate cells. Autophagy. 2018;14(12):2083–103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Zhang J, Wang H, Chen H, Li H, Xu P, Liu B, Zhang Q, Lv C, Song X. ATF3 -activated accelerating effect of LINC00941/lncIAPF on fibroblast-to-myofibroblast differentiation by blocking autophagy depending on ELAVL1/HuR in pulmonary fibrosis. Autophagy. 2022;18(11):2636–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wang JY, Lu AQ. The biological function of m6A reader YTHDF2 and its role in human disease. Cancer Cell Int. 2021;21(1):109.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Chen X, Zhou X, Wang X. mA binding protein YTHDF2 in cancer. Experimental Hematol Oncol. 2022;11(1):21.

    Article  Google Scholar 

  66. Veerappan A, Reid AC, O’Connor N, Mora R, Brazin JA, Estephan R, Kameue T, Chen J, Felsen D, Seshan SV, et al. Mast cells are required for the development of renal fibrosis in the rodent unilateral ureteral obstruction model. Am J Physiol Renal Physiol. 2012;302(1):F192–F204.

    Article  CAS  PubMed  Google Scholar 

  67. Hirooka Y, Nozaki Y, Niki K, Inoue A, Sugiyama M, Kinoshita K, Funauchi M, Matsumura I. Foxp3-Positive Regulatory T cells contribute to Antifibrotic Effects in Renal Fibrosis via an Interleukin-18 receptor signaling pathway. Front Med. 2020;7:604656.

    Article  Google Scholar 

  68. Wang YY, Jiang H, Pan J, Huang XR, Wang YC, Huang HF, To KF, Nikolic-Paterson DJ, Lan HY, Chen JH. Macrophage-to-myofibroblast transition contributes to interstitial fibrosis in Chronic Renal Allograft Injury. J Am Soc Nephrology: JASN. 2017;28(7):2053–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Wilson SE. TGF beta – 1, -2 and – 3 in the modulation of fibrosis in the cornea and other organs. Exp Eye Res. 2021;207:108594.

    Article  CAS  PubMed  Google Scholar 

  70. Lanvin O, Guglielmi P, Fuentes V, Gouilleux-Gruart V, Mazière C, Bissac E, Regnier A, Benlagha K, Gouilleux F, Lassoued K. TGF-beta1 modulates Fas (APO-1/CD95)-mediated apoptosis of human pre-B cell lines. Eur J Immunol. 2003;33(5):1372–81.

    Article  CAS  PubMed  Google Scholar 

  71. Xu A, Liu Y, Chen W, Wang J, Xue Y, Huang F, Rong L, Lin J, Liu D, Yan M, et al. TGF-β-Induced Regulatory T cells directly suppress B cell responses through a noncytotoxic mechanism. J Immunol (Baltimore Md : 1950). 2016;196(9):3631–41.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge the GEO database for providing their platforms and contributors for uploading their datasets.

Funding

No funding.

Author information

Authors and Affiliations

Authors

Contributions

Hanchao Zhang and Guobiao Liang conceived and designed the project. Hanchao Zhang wrote the manuscript. Yue Yang, Zhengdao Liu and Hong Xu analyzed the data. Han Zhu and Peirui Wang helped prepare figures and/or tables. Guobiao Liang critical revision of the manuscript for important intellectual content. All authors contributed to the manuscript and approved the submitted version.

Corresponding author

Correspondence to Guobiao Liang.

Ethics declarations

Conflicts of interest

The authors declare that they have no competing interests.

Ethics approval and consent to participate

Not applicable.

Additional information

Publisher’s Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

41065_2023_295_MOESM1_ESM.png

Additional file 1: Supplementary Figure 1. (A-G) Consensus matrices of the 5 MRGs for k = 3-9. Supplementary Figure 2. (A-G) Consensus matrices of the 56 DEGs for k = 3-9.

41065_2023_295_MOESM2_ESM.png

Additional file 2:Supplementary Table 1. The 48 MRGs. Supplementary Table 2. The 33 differentially expressed MRGs. Supplementary Table 3. The GO enrichment analysis. Supplementary Table 4. The KEGG enrichment analysis.

Supplementary Material 3

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, H., Yang, Y., Liu, Z. et al. Significance of methylation-related genes in diagnosis and subtype classification of renal interstitial fibrosis. Hereditas 160, 32 (2023). https://doi.org/10.1186/s41065-023-00295-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41065-023-00295-8

Keywords