Your privacy, your choice

We use essential cookies to make sure the site can function. We also use optional cookies for advertising, personalisation of content, usage analysis, and social media.

By accepting optional cookies, you consent to the processing of your personal data - including transfers to third parties. Some third parties are outside of the European Economic Area, with varying standards of data protection.

See our privacy policy for more information on the use of your personal data.

for further information and to change your choices.

Skip to main content

Inhibition of complement system-related gene ITGB2 attenuates epithelial–mesenchymal transition and inflammation in diabetic nephropathy

Abstract

Purpose

Emerging evidences have indicated a role of the complement system in the pathogenesis of diabetic nephropathy (DN). Thus, this study was conducted to explore the complement system-related key biomarkers for patients with DN.

Methods

DN microarray datasets were downloaded from the GEO database, followed by differentially expressed genes (DEGs) screening. Complement system-related genes (CSRGs) were searched from various databases. Weighted Gene Co-expression Network Analysis (WGCNA) was employed to screen the DN-related genes, then the differential CSRGs (DCSRGs) were identified, followed by protein–protein interaction (PPI) network construction. In addition, key biomarkers were acquired by two machine learning algorithms, then immune infiltration analysis, Gene Set Enrichment Analysis (GSEA), and potential drugs screening were conducted. Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blotting were utilized to detect the ITGB2 expression. Then the cell viability, inflammatory factors, and the expression of epithelial–mesenchymal transition (EMT) and fibrosis markers were determined by using Cell Counting Kit-8 (CCK-8) assay, enzyme linked immunosorbent assay (ELISA), western blotting assays, respectively.

Results

In total, 1012 DEGs and 974 DN-related genes were screened, and intersection analysis of the three (DN-related genes, DEGs and CSRGs) yielded 13 intersection genes, which were considered as the DCSRGs. Subsequently, 2 key biomarkers were identified by machine learning, namely VWF and ITGB2. The VWF and ITGB2 were both enriched in the pathways of chemokine signaling pathway, CAMs, focal adhesion and natural killer cell-mediated cytotoxicity, and significantly correlated with the activated mast cells, resting NK cells, and macrophages. Also, VWF and ITGB2 were significantly related to the clinical features, including age, serum creatinine level, and GFR (MDRD). Besides, mRNA and protein expression levels of ITGB2 in HG-treated HK-2 cells were remarkably elevated. Moreover, the viability of HK-2 cells, expression of TNF-α, IL-6, IL-12, α-SMA, E-cadherin and vimentin in HK-2 cells changed by HG administration were reversed by ITGB2-silence.

Conclusion

Complement system-related gene ITGB2 was overexpressed in DN, and inhibition of ITGB2 attenuated EMT and inflammation in DN.

Introduction

Diabetic nephropathy (DN) is not only a frequent complication of type 1 and type 2 diabetes mellitus [1], but also one of the most important kidney diseases resulting in end-stage renal disease, which has risen to be the first reason of end-stage renal disease worldwide [2, 3]. According to the International Diabetes Federation, the number of diabetes mellitus patients will increase to 700 million by 2045, and the prevalence of DN, as a common complication of diabetes mellitus, is also increasing year by year [4]. Multiple studies have shown that patients with DN have a significantly increased risk of adverse cardiovascular events, infection and death, which greatly increases socio-economic costs and imposes a heavy economic burden on individuals, families and society [5, 6]. At present, the methods for treating DN are limited, mainly through long-term use of sodium-dependent glucose transporter 2 and renin–angiotensin–aldosterone system inhibitors to control blood glucose and blood pressure, alleviate albuminuria, and delay the kidney disease progression [7, 8]. However, studies have shown that these drugs can only delay but not completely prevent the progression of DN, and most patients with DN will eventually develop end-stage renal disease, and their clinical prognosis is not optimistic [9, 10]. Thus, it is necessary to find effective biomarkers for diagnosing and providing effective therapeutic strategies for DN.

The complement system is the immune response system of the body, which is composed of more than 30 kinds of proteins widely existing in serum, interstitial fluid and cell membrane surface [11, 12]. Complement system, as a significant component of the innate immune system, is the first barrier to accelerate inflammatory responses by improving damaged cells and removing the body’s pathogenic microorganisms [13, 14]. Studies have revealed that the occurrence and development of DN are strongly linked to metabolic abnormalities, innate immunity, and chronic inflammation, etc. [15, 16], and complement system is participated in the progression of a range of diseases, including DN [17,18,19]. Mannan-binding lectin (MBL) is a component of the complement system, and study shows that there is a close relationship between MBL level and microalbuminuria in patients with DN [20], which may be related to the change of glycosylation that can deactivate important complement regulatory proteins on the cells surface, leading to abnormal deposition and MBL activation in the kidney. However, how complement system participated in DN pathogenesis remains unclear.

In this study, the key biomarkers for patients with DN were identified using the Weighted Gene Co-expression Network Analysis (WGCNA) and machine learning, as well as verified in vitro. This study will lay the foundation for the diagnosis and treatment of DN.

Materials and methods

Data source and preliminary processing

Gene expression profile datasets GSE30528 (9 DN patients and 13 healthy controls), GSE30529 (10 DN patients and 12 healthy controls) and GSE104954 (7 DN patients and 18 healthy controls) were acquired from GEO database. The “sva” package [21] (v 3.50.0) was used to merge the GSE30528 and GSE30529 datasets as the training set, and GSE104954 was used as the validation set in this study. Besides, complement system-related genes (CSRGs) were searched from Human Biological Pathway Unification (https://pathcards.genecards.org/), HGNC database [22], and MSigDB database [23] with the keyword of “complement”, and a total of 241 CSRGs were obtained (Supplement file 1).

Identification of differentially expressed genes (DEGs)

The “limma” package [24] (v 3.58.1) was employed to screen DEGs between DN and control samples. Benjamini and Hochberg (BH) method was applied to correct the P value, and the P.adj was obtained. The DEGs were screened with the cutoff value of P.adj < 0.05 and |log2 fold change (FC)|> 0.5.

Weighted Gene Co-expression Network Analysis (WGCNA)

The WGCNA method was applied to identify the DN-related genes. The genes with median absolute deviation (MAD) in the 50% were removed after determining the MAD of each gene. The input genes were analyzed by “WGCNA” package [25] (v 1.72–5) to obtain the key modules. Firstly, square value of related coefficient and average connectivity between the connectivity (k) and p(k) for each power value were calculated by setting a series of powers, and then the optimum power value was chosen to confirm that the connections between genes in the network follow a scale-free network distribution. Secondly, the parameter was set to minModulus Size = 100 to cluster highly associated genes into modules by using clustering and dynamic pruning methods. Then modules strongly associated to DN were identified as key modules by calculating the association between the phenotype (DN) and module with the cutoff value of and P < 0.05. The genes in key modules were considered as the DN-related genes. Then the DN-related genes were intersected with DEGs and CSRGs, and differential CSRGs (DCSRGs) were obtained. Subsequently, “clusterProfiler” (v 4.4.4) was used to conduct enrichment analysis on these DCSRGs with the cutoff value of P.adj < 0.05.

Protein–protein interaction (PPI) network construction

STRING database [26] was employed to search the protein interactions of the DCSRGs with the threshold of score > 0.4, then the PPI network was established.

Identification of key biomarkers

“LASSO” and “Best Subsets Regression (BSR)”, were utilized to screen feature genes based on the obtained DCSRGs. “LASSO” and “BSR” were conducted using “glmnet” [27] (v 4.1–8) and “leaps” [28] (v 3.1), respectively. The “LASSO” was performed with the parameters of random.seed = 12345, nfolds = 10, family = “binomial”, and the parameter in “BSR” was set to random.seed = 2025. Then the overlapping feature genes were considered as the key biomarkers. Moreover, the key biomarkers expression levels between DN and control samples were validated in validation dataset. In addition, the correlation between key biomarkers and clinical features was explored by Nephroseq database [29].

Immune infiltration analysis

“CIBERSORT” algorithm [30] was applied to calculate the fraction of 22 immune cells in samples, and Wilcoxon test was applied to explore the difference of the fraction of immune cells between DN and control samples. The correlation between key biomarkers and immune cells were explored.

GSEA

To further explore the relevant signaling pathways and potential biological mechanisms involved in key biomarkers, correlation analysis was conducted between key biomarkers and all other genes. Then the correlation coefficients were sorted, and GSEA [31] was conducted on the key biomarkers with the cutoff value of P.adj < 0.05 and | Normalized Enrichment Score (NES)|> 1.

Potential drug screening

The DSigDB was employed to analyze the interactions between key biomarkers and related drugs by “enrichR” package (v 3.2) [32]. Then “Cytoscape” software [33] was applied to visualize the key biomarkers-drug network.

Cell culture and transfection

HK-2 cells (CL-0109) purchased from Pricella (Wuhan, China) were maintained in DMEM medium containing normal glucose (5.56 mmol/L) and high glucose (HG; 30 mmol/L) [34] for 24 h in DMEM medium with conventional cultivation conditions. Herein, HG was applied to induce the DN cell model. The loss-of-function assays was performed utilizing lentivirus infection to explore the role of ITGB2 in vitro. For transfection, the sequences for the sh-ITGB2 design were obtained from the Designer of Small Interfering RNA website. ITGB2 shRNA, sense: 5’-GGTGAAGACCTACGAGAAA-3’; antisense: 5’-TTTCTCGTAGGTCTTCACC-3’. The HK-2 cells were transfected with lentivirus containing either sh-ITGB2 or its control (sh-NC) for a duration of 24 h, then exposed to DMEM medium containing HG for 24 h. In brief, the cells were categorized into four groups: control, HG, HG + sh-NC, and HG + sh-ITGB2. For cell viability, a commercial CCK-8 kit (C0037, Beyotime, Shanghai, China) was applied to explore.

Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR)

The qRT-PCR process was carried out in accordance with methods outlined earlier [35]. After treatment, the total RNA was separated using Trizol (15,596,018, Invitrogen, USA) and reverse-transcribed into cDNA by FastKing-RT SuperMix (KR118-02; TIANGEN, Beijing, China). Then qRT-PCR was conducted with SYBR Green PCR Master mix (A4004M, Lifeint, China). The primers used were ITGB2 (Human) forward 5’-GATGACGGCTTCCATTTCGC-3’ and reverse 5’-TGGGGATGATCTCGGTGAGT-3’; GAPDH (Human) forward 5’-CCATGGGGAAGGTGAAGGTC-3’, and reverse 5’-AGTGATGGCATGGACTGTGG −3’. GAPDH was employed as an internal control.

Western blotting analysis

Cells were lysed utilizing RIPA buffer (ST506; Beyotime, Shanghai, China) and BCA protein assay kit (P0010S; Beyotime, Shanghai, China) was applied to detect the protein concentration. Then the obtained proteins were isolated utilizing SDS-PAGE and transferred to PVDF membrane (FFP24; Beyotime, Shanghai, China). Subsequently, the membranes were probed with the antibodies against ITGB2 (1:1000; #DF6896, Affinity, USA), E-cadherin (1:1000; #4065, CST, USA), vimentin (1:1000; #5741, CST, USA), α-SMA (1:1000; #14,968, CST, USA), and GAPDH (1:1000; ab181602, Abcam, USA), followed by secondary antibody goat anti-rabbit IgG H&L (HRP) (1:10,000; ab6721, Abcam, USA). The results were assessed by electrochemical luminescence (ECL) reagents (P1000, APPLYGEN, China).

Enzyme linked immunosorbent assay (ELISA)

The cell supernatant was obtained for ELISA assay. The TNF-α (ml064303; Mlbio, Shanghai, China), IL-6 (ml058097; Mlbio, Shanghai, China), and IL-12 (GOY-1653, Goybio, Shanghai, China) were evaluated with ELISA kits.

Statistical analysis

All data were presented as mean ± standard deviation (SD). Differences between groups were evaluated using one-way ANOVA by GraphPad Prism 7.0 software, followed by Tukey’s post hoc test for comparison. P < 0.05 was indicated statistically significant.

Results

DEGs screening between DN and controls

The before and after removing batch effect of datasets are shown in Fig. 1A and B, respectively. After differential expression analysis, totally 1012 DEGs were acquired, including 613 up- and 399 down-regulated DEGs (Fig. 1C and D). In addition, the CSRG score showed significant difference between DN and controls groups (Fig. 1E), suggesting that CSRG exerted a significant function in the occurrence of DN.

Fig. 1
figure 1

Differentially expressed genes (DEGs) screening between diabetic nephropathy (DN) and controls. Before (A) and after (B) removing batch effect of datasets. Volcano plot (C) and heatmap (D) of top 50 DEGs between DN and control samples. E The difference of complement system-related genes (CSRGs) score between DN and control groups

Identification of DCSRGs

WGCNA was applied to screen the DN-related genes. The scale-free fit index was set to 0.82 to acquire the minimum “soft” threshold of 12 (Fig. 2A). After calculating the adjacency and dissimilarity coefficients between genes (MEDissThres = 0.3), 10 modules were combined (Fig. 2B) when the parameter set to minModulus Size = 100, and blue module was strongly related to DN (Fig. 2C). Then a total of 974 genes in blue module were considered as the DN-related genes. Subsequently, total 13 overlapping genes were obtained in DN-related genes, DEGs and CSRGs, namely VWF, ITGB2, C1QB, C7, VSIG4, FCN1, F12, KNG1, C1S, C3AR1, C3, C1QA, and C1RL, which were considered as the DCSRGs (Fig. 2D).

Fig. 2
figure 2

Identification of differential complement system-related genes (DCSRGs). A Left: scale-free fit index (proportional independence, y-axis) as a function of soft threshold power (x-axis); right: mean connectivity (degrees, y-axis) as a function of soft threshold power (x-axis). B Gene dendrogram as a result of clustering, where the colored rows below the dendrogram denote the module assignment identified by dynamic tree shearing. C Module–DN correlations. D Venn diagram of the DCSRGs

Enrichment analysis and PPI network establishment

Enrichment analysis was conducted to investigate the biological function of DCSRGs. GO analysis revealed that these DCSRGs enriched in 234 GO terms, mainly involving in complement activation, humoral immune response, and others complement/immune-related functions (Fig. 3A). Also, KEGG pathways results demonstrated that the enriched pathways largely involved complement and coagulation cascades, Staphylococcus aureus infection, and systemic lupus erythematosus (Fig. 3B). Besides, PPI network of the DCSRGs were established (Fig. 3C), containing 13 nodes and 46 edges.

Fig. 3
figure 3

Enrichment analysis and protein–protein interaction (PPI) network establishment. Top 10 Gene Ontology (GO) terms (A) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (B) of 13 differential complement system-related genes (DCSRGs) enriched. Among the enriched GO terms, blue bars represent Biological Process (BP) terms, green bars represent Cell Component (CC) terms, and orange bars represent Molecular Function (MF) terms. C Protein–protein interaction (PPI) network. Blue nodes indicate downregulated genes, and red nodes indicate upregulated genes

Key biomarkers screening based on machine learning algorithms

Machine learning has been used in bioinformatics, providing insights into their role in enhancing our understanding of human diseases [36,37,38]. Two machine learning algorithms were utilized to identify key biomarkers. In total, 6 (VWF, ITGB2, C7, VSIG4, KNG1, C3) and 3 (VWF, C1QB, ITGB2) feature genes were obtained by “LASSO” and “BSR” algorithms, respectively (Fig. 4A and B). Notably, 2 common genes were obtained (Fig. 4C), namely VWF and ITGB2, which were considered as the key biomarkers. The AUCs of key biomarkers were all above 0.85 in training set (Fig. 4D), as well as above 0.75 in validation set (Fig. 4E). Furthermore, the VWF and ITGB2 expression levels both remarkably elevated in DN group when compared to those in control group both in training and validation sets (Fig. 4F and G). Also, VWF and ITGB2 were significantly related to the clinical features, including age, serum creatinine level, and GFR(MDRD) (Fig. 4H).

Fig. 4
figure 4

Key biomarkers screening based on machine learning algorithms. Feature genes were obtained using “LASSO” (A) and “BSR” (B). C Venn diagram of key biomarkers. Receiver operating characteristic (ROC) curve of key biomarkers in training (D), and verification (E) sets. The expression levels of key biomarkers between DN and control groups in training (F) and validation (G) sets. *P < 0.05, ****P < 0.0001. H The correlation between key biomarkers and clinical features explored by Nephroseq database

GSEA, immune infiltration analysis and potential drug screening

GSEA uncovered that VWF and ITGB2 were largely involved in the pathways of chemokine signaling pathway, cell adhesion molecules (CAMs), focal adhesion and natural killer cell-mediated cytotoxicity (Fig. 5A and B). In addition, the relative fraction of immune cells in DN and controls is illustrated in Fig. 5C, and activated mast cells, macrophages, resting NK cells showed significantly difference between DN and controls samples (Fig. 5D). Notably, VWF and ITGB2 were significantly correlated with the activated mast cells, macrophages, and resting NK cells (Fig. 5E). Moreover, in total 98 potential drugs related to VWF and ITGB2 were searched, then the key biomarkers–drug network was constructed (Fig. 5F), including 100 nodes and 107 edges.

Fig. 5
figure 5

Gene Set Enrichment Analysis (GSEA), immune infiltration analysis and potential drug screening. GSEA analysis of VWF (A) and ITGB2 (B). C The relative proportion of immune cells in diabetic nephropathy (DN) and controls. D The fraction of immune cells shown significant difference between DN and normal samples. E The correlation between 2 key biomarkers and immune cells. F Drug–key biomarkers network. Red nodes indicate key biomarkers, and yellow nodes indicate drugs

Influence of ITGB2 on viability, EMT, fibrosis, and inflammation in HG-treated HK-2 cells

After bioinformatics analysis, the results found two key biomarkers, namely VWF and ITGB2. It is reported that the VWF expression level was elevated in DN, and inhibition of VWF could improve kidney function in diabetes mice [39]. However, no studies have reported the influence of ITGB2 in DN. Thus, the influences of ITGB2 in DN were explored. The ITGB2 mRNA and protein expression levels in HG-treated HK-2 cells were remarkably elevated (P < 0.01; Fig. 6A–C). Considering the overexpression expression of ITGB2 in HG-treated HK-2 cells, the HG-treated HK-2 cells were transfected with sh-ITGB2, and the transfection efficiency was measured (Fig. 6B–C). Then the viability of cells was explored by CCK-8, and the CCK-8 result showed that the HK-2 cells viability was significantly attenuated by HG compared to control group, and it was significantly increased after inhibition of ITGB2 (both P < 0.05; Fig. 6D). In addition, epithelial–mesenchymal transition (EMT) markers, containing E-cadherin and vimentin were analyzed by western blotting, which illustrated that EMT was significantly increased after HG treatment, and it was significantly mitigated by sh-ITGB2 implement (both P < 0.01), as well as the protein level of fibrosis marker α-SMA (Fig. 6E). Also, TNF-α, IL-6, and IL-12 were detected, which indicated that inflammation was significantly enhanced after HG administration, while it was reverse by ITGB2 knockdown (most P < 0.01; Fig. 6F). These data collectively suggested the inhibition of ITGB2 attenuated EMT, fibrosis, and inflammation in HG-treated HK-2 cells.

Fig. 6
figure 6

Influence of ITGB2 on viability, epithelial–mesenchymal transition (EMT), fibrosis, and inflammation in HG-treated HK-2 cells. A The mRNA expression levels of ITGB2 in HG-treated HK-2 cells. Transfection efficiency was detected by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) (B) and western blotting analysis (C). D The proliferation of cells was detected using Cell Counting Kit-8 (CCK-8) assay. E EMT and fibrosis markers, containing E-cadherin, vimentin and αSMA were analyzed by western blotting analysis. F TNF-α, IL-6, and IL-12 expression levels detected by enzyme linked immunosorbent assay (ELISA). **P < 0.01, compared with control group; #P < 0.05, ##P < 0.01, compared with HG + sh-NC group

Discussion

DN is one of the major causes of death in diabetes mellitus patients [40]. Numerous studies have suggested that complement system exerts significant functions in the pathogenesis of DN [41,42,43]. Therefore, screening specific complement system-related biomarkers might lead to identification of potential diagnostic and therapeutic targets for DN patients. In this study, two complement system-related key biomarkers were identified by WGCNA and machine learning, namely VWF and ITGB2. In addition, the AUCs of VWF and ITGB2 were all above 0.75 in training and validation sets, indicating good predictive performance. Furthermore, the VWF and ITGB2 expression levels both significantly elevated in DN group when compared to those in control group both in training and validation sets, and VWF and ITGB2 were significantly related to the clinical features. The results of this study are in line with numerous reported studies, which revealed that the level of VWF antigen is elevated in type 1 and type 2 diabetes mellitus patients with DN [39, 44, 45]. In addition, Hu et al. using bioinformatics analysis found that ITGB2 was upregulated in DN samples, which could be used as a underlying diagnostic gene for DN [46], as well as the study reported by Xu et al. [29], which were further confirmed that VWF and ITGB2 might viable biomarkers in DN. Besides, GSEA uncovered that VWF and ITGB2 were largely involved in the pathways of chemokine signaling pathway, CAMs, focal adhesion and natural killer cell-mediated cytotoxicity. Thus, VWF and ITGB2 might exert vital functions in the occurrence and development of DN through chemokine signaling pathway, CAMs, focal adhesion and natural killer cell-mediated cytotoxicity pathway.

As we all know, immunity is one of the key mechanisms in the DN progression. Hyperglycemia is a key factor contributing to kidney damage in patients with DN [47]. The stress caused by sustained hyperglycemia can lead to the massive production of inflammatory molecules and the immune complexes accumulation, which is strongly associated to immune cells such as NK cells, mast cells, and T cells [48]. The results of immune infiltration analysis in this study also indicated that VWF and ITGB2 were significantly correlated with the activated mast cells, macrophages, and resting NK cells. It is also reported that macrophages are the main innate immune cells in DN, and their presence in interstitium and glomeruli can be observed in both clinical trials and experimental models of DN [49, 50]. Thus, VWF and ITGB2 might the therapeutic targets for DN immunotherapy. Moreover, in total 98 potential drugs related to VWF and ITGB2 were searched. Among which, progesterone CTD 00006624, ACMC-20mvek CTD 00002629, and 9001–31-4 BOSS, etc., both targeted VWF and ITGB2, which suggesting that progesterone CTD 00006624, ACMC-20mvek CTD 00002629, and 9001–31-4 BOSS, etc., that targeted VWF and ITGB2 might have therapeutic potential for DN.

Besides, this study further verified the key biomarkers in vitro. However, emerging evidences have revealed a role for VWF in the pathogenesis of DN; no studies have reported the specific influence of ITGB2 in DN. Thus, the influences of ITGB2 in DN were explored in depth in this study. HG is a recognized important risk factor for DN and is closely related to the pathology and progression of DN [51, 52]. Therefore, HK-2 cells were maintained in HG (30 mM) medium to build a cellular model of DN [53]. Consistent with the results of bioinformatics analysis, ITGB2 mRNA and protein expression levels in HG-treated HK-2 cells were remarkably elevated. EMT exerts significant functions in interstitial matrix deposition in DN [54]. Up to 30–40% of type 1 or type 2 diabetes mellitus patients will develop to DN with characterized by of ECM protein accumulation [55, 56]. The enhanced ECM deposition then leads to renal fibrosis, ultimately resulting in tubulointerstitial fibrosis, infiltration of inflammatory mediators, and reactivation of α-SMA-positive myofibroblasts [57, 58]. In this study, the viability of HK-2 cells, expression of inflammatory regulators, fibrosis marker α-SMA, and EMT markers in HK-2 cells changed by HG administration were reversed by ITGB2-silence. Taken together, these data suggested that inhibition of ITGB2 alleviated cell viability, EMT, fibrosis, and inflammation in HG-treated HK-2 cells. In addition, the US-FDA have supported numerous siRNA delivery systems for various diseases treatment [59,60,61], whether nanoparticle loading siITGB2 could be used for clinical therapy of DN need further investigated.

This study still has numerous limitations. Firstly, the 241 CSRGs were obtained from Human Biological Pathway Unification, HGNC, and MSigDB databases, which is being updated continuously, and more CSRGs should be discovered. In addition, the molecular mechanisms of ITGB2 in DN should be further explored. Lastly, whether the identified drugs that targeted VWF and ITGB2 could be suitable for clinical application in DN treatment also need to be deeply investigated at the clinical level.

Conclusion

In conclusion, complement system-related gene ITGB2 was overexpressed in DN, which was a potential diagnostic and therapeutic target in DN. In addition, inhibition of ITGB2 mitigated EMT and inflammation in DN. This study offers novel insights for DN diagnosis and treatment.

Availability of data and materials

The raw data of bioinformatics analyses of current study are openly available in public databases listed within the article. The raw data of experiments are available on reasonable requests.

References

  1. Kriz W, Löwen J, Gröne HJ. The complex pathology of diabetic nephropathy in humans. Nephrol Dial Transplan. 2023;38(10):2109–19.

    Article  CAS  Google Scholar 

  2. Pan W, Zhang Q, Gong X, Wu W, Zhou Q. Identification and validation of key extracellular proteins as the potential biomarkers in diabetic nephropathy. Eur J Med Res. 2024;29(1):517.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Liu M, Zhao J. Circular RNAs in diabetic nephropathy: updates and perspectives. Aging Dis. 2022;13(5):1365–80.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the international diabetes federation diabetes Atlas, 9(th) edition. Diabetes Res Clin Pract. 2019;157: 107843.

    Article  PubMed  Google Scholar 

  5. Tuttle KR, Cherney DZI. Therapeutic transformation for diabetic kidney disease. Kidney Int. 2021;99(2):301–3.

    Article  PubMed  CAS  Google Scholar 

  6. Wheeler DC, Stefánsson BV, Jongs N, Chertow GM, Greene T, Hou FF, et al. Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol. 2021;9(1):22–31.

    Article  PubMed  CAS  Google Scholar 

  7. Alder H, Ambühl PM. Managing diabetic kidney disease. Therapeutische Umschau Revue therapeutique. 2020;77(7):333–8.

    Article  PubMed  Google Scholar 

  8. Li Y, Pang S, Guo H, Yang S. The renal apical sodium-dependent bile acid transporter expression rescue attenuates renal damage in diabetic nephropathy via farnesoid X receptor activation. Nephrology. 2024. https://doi.org/10.1111/nep.14338.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Albvr VR, Tan SH, Candasamy M, Bhattamisra SK. Diabetic nephropathy: an update on pathogenesis and drug development. Diabet Metabolic Syndrome. 2019;13(1):754–62.

    Article  Google Scholar 

  10. Kushwaha K, Kabra U, Dubey R, Gupta J. Diabetic nephropathy: pathogenesis to Cure. Curr Drug Targets. 2022;23(15):1418–29.

    Article  PubMed  Google Scholar 

  11. Lasorsa F, Rutigliano M, Milella M, Ferro M, Pandolfo SD, Crocetto F, et al. Complement system and the kidney: its role in renal diseases, kidney transplantation and renal cell carcinoma. Int J Mol Sci. 2023. https://doi.org/10.3390/ijms242216515.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Merle NS, Roumenina LT. The complement system as a target in cancer immunotherapy. Euro J Immunol. 2024. https://doi.org/10.1002/eji.202350820.

    Article  Google Scholar 

  13. Li L, Chen L, Zang J, Tang X, Liu Y, Zhang J, et al. C3a and C5a receptor antagonists ameliorate endothelial–myofibroblast transition via the Wnt/β-catenin signaling pathway in diabetic kidney disease. Metabolism. 2015;64(5):597–610.

    Article  PubMed  CAS  Google Scholar 

  14. Sun ZJ, Li XQ, Chang DY, Wang SX, Liu G, Chen M, et al. Complement deposition on renal histopathology of patients with diabetic nephropathy. Diabetes Metab. 2019;45(4):363–8.

    Article  PubMed  CAS  Google Scholar 

  15. Gupta S, Dominguez M, Golestaneh L. Diabetic kidney disease: an update. Med Clin North Am. 2023;107(4):689–705.

    Article  PubMed  Google Scholar 

  16. Tang SCW, Yiu WH. Innate immunity in diabetic kidney disease. Nat Rev Nephrol. 2020;16(4):206–22.

    Article  PubMed  CAS  Google Scholar 

  17. Jensen M, Eickhoff MK, Persson F, Rossing P, Thiel S, Hansen SWK, et al. Effect of dapagliflozin on collectins and complement activation in plasma from patients with type 2 diabetes and albuminuria: data from the DapKid cohort. Immunobiology. 2024;229(3): 152797.

    Article  PubMed  CAS  Google Scholar 

  18. Jiang F, Lei C, Chen Y, Zhou N, Zhang M. The complement system and diabetic retinopathy. Surv Ophthalmol. 2024;69(4):575–84.

    PubMed  Google Scholar 

  19. Ong J, Zarnegar A, Selvam A, Driban M, Chhablani J. The complement system as a therapeutic target in retinal disease. Medicina. 2024. https://doi.org/10.3390/medicina60060945.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Hovind P, Hansen TK, Tarnow L, Thiel S, Steffensen R, Flyvbjerg A, et al. Mannose-binding lectin as a predictor of microalbuminuria in type 1 diabetes: an inception cohort study. Diabetes. 2005;54(5):1523–7.

    Article  PubMed  CAS  Google Scholar 

  21. 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 (Oxford, England). 2012;28(6):882–3.

    PubMed  CAS  Google Scholar 

  22. Seal RL, Braschi B, Gray K, Jones TEM, Tweedie S, Haim-Vilmovsky L, et al. Genenames.org: the HGNC resources in 2023. Nucleic Acids Res. 2023;51(1):1003.

    Article  Google Scholar 

  23. Castanza AS, Recla JM, Eby D, Thorvaldsdóttir H, Bult CJ, Mesirov JP. Extending support for mouse data in the molecular signatures database (MSigDB). Nat Methods. 2023;20(11):1619–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Liu S, Wang Z, Zhu R, Wang F, Cheng Y, Liu Y. Three differential expression analysis methods for RNA sequencing: limma, EdgeR, DESeq2. J Visualiz Exp. 2021. https://doi.org/10.3791/62528-v.

    Article  Google Scholar 

  25. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics. 2008;9:559.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Szklarczyk D, Kirsch R, Koutrouli M, Nastou K, Mehryary F, Hachilif R, et al. The STRING database in 2023: protein–protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 2023;51(D1):D638–46.

    Article  PubMed  CAS  Google Scholar 

  27. Liu Y, Jiang Z, Zhou X, Li Y, Liu P, Chen Y, et al. A multi-omics analysis of NASH-related prognostic biomarkers associated with drug sensitivity and immune infiltration in hepatocellular carcinoma. J Clin Med. 2023. https://doi.org/10.3390/jcm12041286.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Settino M, Cannataro M. Using MMRFBiolinks R-package for discovering prognostic markers in multiple Myeloma. Methods Mol Biol. 2022;2401:289–314.

    Article  PubMed  CAS  Google Scholar 

  29. Xu M, Zhou H, Hu P, Pan Y, Wang S, Liu L, et al. Identification and validation of immune and oxidative stress-related diagnostic markers for diabetic nephropathy by WGCNA and machine learning. Front Immunol. 2023;14:1084531.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Chen B, Khodadoust MS, Liu CL, Newman AM, Alizadeh AA. Profiling tumor infiltrating immune cells with CIBERSORT. Methods Mol Biol (Clifton, NJ). 2018;1711:243–59.

    Article  CAS  Google Scholar 

  31. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102(43):15545–50.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Pereira EPV, da Silva Felipe SM, de Freitas RM, da Cruz Freire JE, Oliveira AER, Canabrava N, et al. Transcriptional profiling of SARS-CoV-2-infected calu-3 cells reveals immune-related signaling pathways. Pathogens. 2023;12:11.

    Article  Google Scholar 

  33. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Liu Y, Zhang M, Zeng L, Lai Y, Wu S, Su X. Wogonin upregulates SOCS3 to alleviate the injury in diabetic nephropathy by inhibiting TLR4-mediated JAK/STAT/AIM2 signaling pathway. Mol Med. 2024;30(1):78.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Sindhuja S, Amuthalakshmi S, Nalini CN. A review on PCR and POC-PCR - a boon in the diagnosis of COVID-19. Curr Pharm Anal. 2022;18(8):745–64.

    Article  CAS  Google Scholar 

  36. Chattopadhyay SJM. Decoding medical diagnosis with machine learning classifiers. Medinformatics. 2024. https://doi.org/10.47852/bonviewMEDIN42022583.

    Article  Google Scholar 

  37. Mamchur A, Sharashkina N, Erema V, Kashtanova D, Ivanov M, Bruttan M, et al. Machine learning-based decision-making in geriatrics: aging phenotype calculator and survival prognosis. Aging and disease. 2024.

  38. Singh AK, Ling J, Malviya R. Prediction of cancer treatment using advancements in machine learning. Recent Pat Anti-Cancer Drug Discovery. 2023;18(3):364–78.

    Article  PubMed  CAS  Google Scholar 

  39. Dhanesha N, Doddapattar P, Chorawala MR, Nayak MK, Kokame K, Staber JM, et al. ADAMTS13 retards progression of diabetic nephropathy by inhibiting intrarenal thrombosis in mice. Arterioscler Thromb Vasc Biol. 2017;37(7):1332–8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Wu Q, Huang F. Targeting ferroptosis as a prospective therapeutic approach for diabetic nephropathy. Ann Med. 2024;56(1):2346543.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Flyvbjerg A. The role of the complement system in diabetic nephropathy. Nat Rev Nephrol. 2017;13(5):311–8.

    Article  PubMed  CAS  Google Scholar 

  42. Xu B, Wang L, Zhan H, Zhao L, Wang Y, Shen M, et al. Investigation of the mechanism of complement system in diabetic nephropathy via bioinformatics analysis. J Diabetes Res. 2021;2021:5546199.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Li XQ, Chang DY, Chen M, Zhao MH. Complement activation in patients with diabetic nephropathy. Diabetes Metab. 2019;45(3):248–53.

    Article  PubMed  CAS  Google Scholar 

  44. Domingueti CP, Dusse LM, Fóscolo RB, Reis JS, Annichino-Bizzacchi JM, Orsi FL, et al. Von willebrand factor, ADAMTS13 and D-Dimer Are correlated with different levels of nephropathy in type 1 diabetes mellitus. PLoS ONE. 2015;10(7): e0132784.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Hirano T, Ookubo K, Kashiwazaki K, Tajima H, Yoshino G, Adachi M. Vascular endothelial markers, von Willebrand factor and thrombomodulin index, are specifically elevated in type 2 diabetic patients with nephropathy: comparison of primary renal disease. Clin Chim Acta. 2000;299(1–2):65–75.

    Article  PubMed  CAS  Google Scholar 

  46. Hu Y, Yu Y, Dong H, Jiang W. Identifying C1QB, ITGAM, and ITGB2 as potential diagnostic candidate genes for diabetic nephropathy using bioinformatics analysis. PeerJ. 2023;11: e15437.

    Article  PubMed  PubMed Central  Google Scholar 

  47. DeFronzo RA, Reeves WB, Awad AS. Pathophysiology of diabetic kidney disease: impact of SGLT2 inhibitors. Nat Rev Nephrol. 2021;17(5):319–34.

    Article  PubMed  CAS  Google Scholar 

  48. Tesch GH. Diabetic nephropathy - is this an immune disorder? Clin Sci. 2017;131(16):2183–99.

    Article  CAS  Google Scholar 

  49. Chow FY, Nikolic-Paterson DJ, Atkins RC, Tesch GH. Macrophages in streptozotocin-induced diabetic nephropathy: potential role in renal fibrosis. Nephrol Dial Transplan. 2004;19(12):2987–96.

    Article  CAS  Google Scholar 

  50. Guiteras R, Sola A, Flaquer M, Manonelles A, Hotter G, Cruzado JM. Exploring macrophage cell therapy on diabetic kidney disease. J Cell Mol Med. 2019;23(2):841–51.

    Article  PubMed  CAS  Google Scholar 

  51. Nakamichi R, Hayashi K, Itoh H. Effects of high glucose and lipotoxicity on diabetic podocytes. Nutrients. 2021. https://doi.org/10.3390/nu13010241.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Yang J, Liu Z. Mechanistic pathogenesis of endothelial dysfunction in diabetic nephropathy and retinopathy. Front Endocrinol. 2022;13: 816400.

    Article  Google Scholar 

  53. Zheng S, Zhao N, Feng C, Ma J. Cell division cycle 42 attenuates high glucose-treated renal tubular epithelial cell apoptosis, fibrosis, and inflammation, but activates the PAK1/AKT pathway. Clin Exp Nephrol. 2024;28(6):513–21.

    Article  PubMed  CAS  Google Scholar 

  54. Loeffler I, Wolf G. Epithelial-to-mesenchymal transition in diabetic nephropathy: fact or fiction? Cells. 2015;4(4):631–52.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Hu C, Sun L, Xiao L, Han Y, Fu X, Xiong X, et al. Insights into the mechanisms involved in the expression and regulation of extracellular matrix proteins in diabetic nephropathy. Curr Med Chem. 2015;22(24):2858–70.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Loeffler I, Liebisch M, Wolf G. Collagen VIII influences epithelial phenotypic changes in experimental diabetic nephropathy. Am J Physiol Renal Physiol. 2012;303(5):F733–45.

    Article  PubMed  CAS  Google Scholar 

  57. Zeisberg M, Neilson EG. Mechanisms of tubulointerstitial fibrosis. J Am Soc Nephrol. 2010;21(11):1819–34.

    Article  PubMed  CAS  Google Scholar 

  58. Hills CE, Squires PE. The role of TGF-β and epithelial-to mesenchymal transition in diabetic nephropathy. Cytokine Growth Factor Rev. 2011;22(3):131–9.

    PubMed  CAS  Google Scholar 

  59. Qin S, Wang Y, Ma C, Lv Q. Competitive endogenous network of circRNA, lncRNA, and miRNA in osteosarcoma chemoresistance. Eur J Med Res. 2023;28(1):354.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Chen Y, Huang Y, Li Q, Luo Z, Zhang Z, Huang H, et al. Targeting Xkr8 via nanoparticle-mediated in situ co-delivery of siRNA and chemotherapy drugs for cancer immunochemotherapy. Nat Nanotechnol. 2023;18(2):193–204.

    Article  PubMed  Google Scholar 

  61. Wu Z, Tang Y, Liu Y, Chen Z, Feng Y, Hu H, et al. Co-delivery of fucoxanthin and Twist siRNA using hydroxyethyl starch-cholesterol self-assembled polymer nanoparticles for triple-negative breast cancer synergistic therapy. J Adv Res. 2024. https://doi.org/10.1016/j.jare.2024.04.017.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

The author(s) reported there is no funding associated with the work featured in this article.

Author information

Authors and Affiliations

Authors

Contributions

Conception and design of the research: Jun Peng; Acquisition of data: Wenqi Zhao, Lu Zhou; Analysis and interpretation of data: Wenqi Zhao, Lu Zhou; Statistical analysis: Wenqi Zhao, Kun Ding; Drafting the manuscript: Jun Peng; Revision of manuscript for important intellectual content: Jun Peng, Wenqi Zhao, Lu Zhou, Kun Ding. All authors reviewed the results and approved the final version of the manuscript.

Corresponding author

Correspondence to Jun Peng.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher's Note

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

Supplementary Information

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Peng, J., Zhao, W., Zhou, L. et al. Inhibition of complement system-related gene ITGB2 attenuates epithelial–mesenchymal transition and inflammation in diabetic nephropathy. Eur J Med Res 30, 87 (2025). https://doi.org/10.1186/s40001-025-02323-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-025-02323-x

Keywords