Next Article in Journal
Brain and Serum Membrane Vesicle (Exosome) Profiles in Experimental Alcohol-Related Brain Degeneration: Forging the Path to Non-Invasive Liquid Biopsy Diagnostics
Previous Article in Journal
Role of Cathelicidins in Atherosclerosis and Associated Cardiovascular Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

In Silico Characterization of Inflammatory and Anti-Inflammatory Modulation in Diabetic Nephropathy: The Construction of a Genetic Panel

by
Caroline Christine Pincela da Costa
1,
Leandro do Prado Assunção
1,
Kamilla de Faria Santos
1,
Laura da Silva
1,
Rodrigo da Silva Santos
1,2 and
Angela Adamski da Silva Reis
1,2,*
1
Laboratory of Molecular Pathology, Institute of Biological Sciences (ICB II), Federal University of Goiás (UFG), Goiânia 74690-900, GO, Brazil
2
Department of Biochemistry and Molecular Biology, Institute of Biological Sciences (ICB II), Federal University of Goiás (UFG), Goiânia 74690-900, GO, Brazil
*
Author to whom correspondence should be addressed.
J. Mol. Pathol. 2024, 5(3), 335-359; https://doi.org/10.3390/jmp5030024
Submission received: 1 July 2024 / Revised: 13 August 2024 / Accepted: 26 August 2024 / Published: 31 August 2024

Abstract

:
Diabetic Nephropathy (DN) stands as a primary cause of end-stage renal disease and its etiology remains unclear. Thus, this study aims to construct a genetic panel with potential biomarkers linked to the inflammatory pathway of DN associated with the pathology’s susceptibility. Through a systematic review and meta-analysis, we selected observational studies in English, Portuguese, and Spanish, selected from the PubMed, SCOPUS, Virtual Health Library, Web of Science, and EMBASE databases. Additionally, a protein–protein interaction network was constructed to list hub genes, with differential expression analysis by microarray of kidneys with DN from the GSE30529 database to further refine results. Seventy-two articles were included, and 54 polymorphisms in 37 genes were associated with the inflammatory pathway of DN. Meta-analysis indicated a higher risk of complication associated with SNPs 59029 G/A, −511 C/T, VNTR 86 bp, −308 G/A, and −1031 T/C. Bioinformatics analyses identified differentially expressed hub genes, underscoring the scarcity of studies on CCL2 and VEGF-A genes in relation to DN. This study highlighted the intrinsic relationship between inflammatory activity in the etiology and progression of DN, enabling the effective application of precision medicine in diabetic patients for potential prognosis of the complications and contributing to cost reduction in the public health system.

1. Introduction

Diabetic Nephropathy (DN), also known as diabetic kidney disease, is considered the leading cause of end-stage renal failure. However, its etiology has not been fully elucidated [1,2]. Characterized by progressive and irreversible loss of renal function due to chronic hyperglycemia, DN results in glomerular lesions leading to a decline in glomerular filtration rate (GFR) and albuminuria [3].
Chronic hyperglycemia increases the expression of inflammatory mediators due to the injury of tubular and glomerular cells, contributing to renal damage through mesangial proliferation, podocytic/tubular damage, and leukocyte infiltration [4,5]. Hemodynamic changes found in DN, which are associated with hyperglycemia, cause glomerular infiltration, mechanical stress, and endothelial activation. Endothelial cells upregulate the expression of adhesion molecules and chemokines involved in leukocyte infiltration into the renal interstitium [6].
Chemokines enable the activation of integrins by leukocytes, resulting in the adhesion of inflammatory cells to the endothelium, followed by transmigration into various tissues [5,7]. After infiltration into inflammatory foci, leukocytes promote kidney damage through interaction and activation of glomerular and tubular cells generated by the release of chemokines, cytokines, and profibrotic factors, activating stromal renal cells that release additional chemokines and promote increased leukocyte infiltration. These mechanisms amplify the inflammatory response in a positive feedback loop that exacerbates renal damage [4,5].
Currently, the diagnosis of DN is conducted through clinical assessment and laboratory tests, such as evaluation of urinary albumin/creatinine ratio (altered values > 30 mg/g) and estimated GFR (altered values < 60 mL/min/1.73 m2). Furthermore, biopsy is the gold standard for DN diagnosis [8]. Being a multifactorial disease, not all factors that cause the disease are known. Studies indicate that DN is influenced by environmental factors such as sedentary lifestyle, hypertension, and chronic hyperglycemia [9]. However, despite lifestyle changes and glycemic control measures reducing the proportion of diabetic patients progressing to DN, these approaches are insufficient to prevent the risk of developing the disease. Epidemiological studies suggest that genetic factors play a significant role in susceptibility to DN [10].
Therefore, the objective of this study was to identify genetic polymorphisms related to the inflammatory pathway of DN through a systematic review (SR) and meta-analysis for the construction of a genetic panel. Additionally, since the identified genes operate in the same pathway and do not act individually, through in silico analysis, we aimed to identify protein–protein interaction (PPI) to classify hub genes and determine if these genes are differentially expressed genes (DEGs) using the GSE30529 dataset. The integration of these data will assist in identifying potential genetic biomarkers related to the pathogenesis and susceptibility of this silent, progressive, and irreversible disease.

2. Materials and Methods

2.1. Systematic Review

To identify the key genetic polymorphisms associated with the inflammatory process in DN, a systematic review (SR) was conducted guided by the research question: “What are the main genetic polymorphisms related to the inflammatory process in the risk of developing Diabetic Nephropathy?” The review was structured according to the PEO acronym (Population, Exposure, Outcome) (Table 1).
To avoid duplications of this SR, this study had its protocol registered in the International Prospective Register of Systematic Reviews (PROSPERO) on 17 February 2021 (registration number CRD42021232347). For a more comprehensive data description, we adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.
The literature search took place between November 2020 and April 2021 across the PubMed, SCOPUS, Virtual Health Library (Biblioteca Virtual em Saúde—BVS), Web of Science, and EMBASE databases. Studies included in the SR were observational studies assessing humans, conducted in English, Portuguese, or Spanish, with no time restrictions. Combined terms cataloged in the Medical Subject Headings (MeSH) for ‘Diabetic Nephropathy’, ‘Genetic Polymorphism’, and ‘Inflammation’ were employed to formulate the search strategy. The search strategy was adapted for all databases (Table 2).
The exclusion criteria were as follows: articles with a different study design, articles in languages other than English, Portuguese, or Spanish, those that did not evaluate DN, articles without a significant association and not addressing genetic polymorphisms, studies involving animals, and those focusing on genes not participating in the inflammatory pathway.
The articles identified in the search were imported into the Rayyan platform [11]. Screening was conducted by two independent reviewers (LS and KFS) and divided into two phases (I and II), where articles were evaluated based on pre-established inclusion and exclusion criteria. In Phase I, titles and abstracts were reviewed, selecting only articles addressing polymorphisms in genes related to the inflammatory pathway in DN. Phase II involved a full reading of the articles selected in Phase I. Bias risk was assessed using The Joanna Briggs Institute’s (JBI’s) critical appraisal tool, and applied for each study design. Only studies that positively answered at least 70% of the questions were considered to have a low risk of bias. Discrepancies between reviewers were discussed and resolved through consensus.
For the construction of the genetic panel, the following data were extracted: (1) authors and year of publication; (2) study design; (3) population; (4) sample size; (5) type of diabetes mellitus (DM); (6) gene and its location; (7) polymorphism(s) assessed; (8) genotypic and allelic frequencies; (9) comparison performed; (10) chi-square value; (11) odds ratio (OR)—95% confidence interval (CI); and (12) p-value. Data extraction were performed independently by the two reviewers for all included studies.

2.2. Meta-Analysis

The association of polymorphisms with DN was estimated by calculating the odds ratio (OR) with a 95% Confidence Interval (CI), using the dominant genetic model (wild type vs. heterozygote + mutant). Heterogeneity among studies was assessed using Higgins’ inconsistency test (I2). The selection of the meta-analytic model was based on the result of the heterogeneity test among studies. Therefore, the fixed-effect model (Mantel–Haenszel method) was applied when I2 < 25% (low heterogeneity), assuming that discrepancies between effect estimates are merely due to chance. The random-effects model (DerSimonian–Laird method) was applied when I2 > 25% (moderate or high heterogeneity).
Publication bias was assessed by a funnel plot [12], and its asymmetry was estimated using linear regression (Egger’s test) [13]. A p-value < 0.05 suggests a high likelihood of publication bias. All statistical analyses were performed using RStudio® software (version 4.1.0).

2.3. Protein–Protein Interaction and Network Analysis

The list of genes from the constructed genetic panel was exported to the Search Tool for the Retrieval of Interacting Genes (STRING) database for the construction of the protein network. Subsequently, Cytoscape software (version 3.6.1, http://www.cytoscape.org/, accessed on 22 November 2023) was employed to visualize the protein–protein interaction (PPI) network, and the identification of the top 20 hub genes was performed using the CytoHubba tool.

2.4. Microarray Data and Data Processing

The microarray expression dataset used was GSE30529, and was obtained from the National Center for Biotechnology Information-Gene Expression Omnibus (NCBI-GEO) public database (https://www.ncbi.nlm.nih.gov/geo/, 13 December 2023). This dataset was registered on the GPL57 platform ([HG-U133A_2] Affymetrix Human Genome U133A 2.0 Array) and provides the gene expression profile of 10 samples from kidneys with DN and 12 control samples.
To identify differentially expressed genes (DEGs), two approaches were employed. The first involved a parametric statistical analysis with p-value correction using the False Discovery Rate (FDR) method proposed by Benjamini and Hochberg, with a significance level of 5%. Subsequently, a fold change of 1.2 was applied to identify proteins that were biologically regulated (up-regulated, down-regulated, and not significant). All analyses were performed using the GEO2R tool (https://www.ncbi.nlm.nih.gov/geo/info/geo2r.html, 13 December 2023) along with the GEOquery and Limma libraries of R software version 4.2.1.

3. Results

3.1. Systematic Review and Meta-Analysis

Following the initial search, 1782 articles were identified in the databases, with 387 duplicates excluded, resulting in 1395 articles for Phase I of the SR. Based on the review of titles and abstracts, all articles addressing polymorphisms in genes related to the inflammatory pathway in DN were selected. Of the initial pool, 1298 articles were excluded according to pre-established exclusion criteria. Phase II involved a thorough reading of the 97 selected articles, resulting in the exclusion of 25 articles and the inclusion of 72 in the SR (Figure 1), published between 1996 and 2020.

3.2. Methodological Quality Analysis

The 72 studies demonstrated consistency in quality assessment, each achieving a minimum of 70% positive responses on the critical appraisal tool of the Joanna Briggs Institute (JBI) (Supplementary File S1) and thus classified as low risk of bias. In case-control studies (Figure 2A), the majority responded positively to most questions, except for question number 6, as expected, given its evaluation of the presence of confounding factors. Regarding cohort studies (Figure 2B), questions 6, 8, 9, and 10 were deemed not applicable. Most articles appropriately addressed the remaining questions, except for questions 4 and 5, which focused on confounding factors and strategies to address them. In cross-sectional studies (Figure 2C), despite being in the minority, they responded positively to the questionnaire.

3.3. Individual Results

Among the included articles, 61 were case-control studies, 9 were cohort studies, and 2 were cross-sectional studies. From the data extraction, 54 polymorphisms in 37 genes associated with the inflammatory pathway of DN were identified (Table 3).
The genes and their respective polymorphisms most frequently mentioned in the systematic review were: CCR5 (59029 G/A), IL1B (−511 C/T), TGF-1B (869 T/C), IL10 (−1082 G/A), IL1RN (VNTR of 86 bp), and TNF (−308 G/A and −1031 T/C). The 59029 G/A polymorphism of the CCR5 gene was assessed in 8 articles in this systematic review. The meta-analysis results indicated an association between this SNP and the risk of DN (OR = 1.88; 95% CI = 1.23–2.88; and p = 0.0035) (Figure 3).
Seven studies were included in the meta-analysis that observed the SNP −511 C/T in the IL1B gene, and the results indicated an association of this SNP with the risk of developing DN (OR = 1.55; 95% CI = 1.12–2.16; and p = 0.0081) (Figure 4).
In the TGF-1B gene, the SNP 869 T/C was identified in six articles. The meta-analysis showed no association with the risk of developing DN (OR = 2.14; 95% CI = 0.95–4.82; and p = 0.0669) (Figure 5). Additionally, the SNP −1082 G/A in the IL-10 gene also showed no association with the risk of developing DN based on the meta-analysis (OR = 1.51; 95% CI = 0.79–2.87; and p = 0.2150) (Figure 6).
We identified in the IL-1RN gene a variable number tandem repeat (VNTR) polymorphism of 86 bp, which was significantly associated with the risk of developing DN (OR = 2.42; 95% CI = 1.03–5.65; and p = 0.0420) (Figure 7). Two polymorphisms in the TNF gene showed a statistically significant risk in the development of DN; namely, −308 G/A (OR = 1.98; 95% CI = 1.33–2.96; and p = 0.0008) (Figure 8) and −1031 T/C (OR = 2.73; 95% CI = 1.76–4.21; and p = < 0.0001) (Figure 9).
The publication bias analysis for the SNPs 59029 G/A, −511 C/T, 869 T/C, −1082 G/A, VNTR of 86 bp, −308 G/A, and −1031 T/C did not show publication bias according to the funnel plot (Figure 10) and Egger’s test (p = 0.8884, p = 0.8938, p = 0.0784, p = 0.7868, p = 0.6703, p = 0.8336, and p = 0.5576, respectively).

3.4. Protein Network Analysis and Identification of Hub Genes

The selected genes participate in the same pathway and do not act in isolation; therefore, we constructed a protein–protein interaction (PPI) network to identify the interactions between the proteins encoded by the selected genes. This network included 34 nodes and 261 edges, where each node represents a protein and each edge represents an interaction between proteins (Figure 11). The PPI enrichment p-value was <1.0 × 10−16, indicating that the protein network had significantly more interactions than expected and is biologically connected.
The analysis of hub genes revealed the top 20 genes with the highest interaction in the inflammatory pathway of DN. These genes are as follows: TNF, IL1B, IL6, CCL2, IL10, ICAM1, CXCL8, MMP9, TLR4, VEGFA, IL4, IL1A, CCL5, MPO, ADIPOQ, TGFB1, LTA, SPP1, IL1RN, and NOS2 (Figure 12 and Table 4).

3.5. Differential Expression Analysis—GSE30529

For the analysis of transcript data, 22,277 genes from the database GSE30529, available at the National Center for Biotechnology Information-Gene Expression Omnibus (NCBI-GEO) public database (https://www.ncbi.nlm.nih.gov/geo/, 13 December 2023), were considered. To screen for the genes with a statistical difference (p-value ≤ 0.05), a hypothesis test was performed considering the FDR, in which 4184 genes were found. Subsequently, a fold-change of 1.2 was considered to identify the DEG. Sixty-two genes were found to be down-regulated, 454 genes were up-regulated, and 3668 genes were not significant (Figure 13) (Supplementary File S2). Among the DEG (down- and up-regulated), three genes were previously identified in the systematic review of the present study: CCL5 (RANTES), CCL2 (MCP-1), and CXCL8 (IL8).

4. Discussion

Despite DN not being classified as an inflammatory disease, numerous studies suggest that molecules within the inflammatory pathway may play significant pathophysiological roles in this complication [84]. Therefore, the primary objective of this study was to identify potential molecular markers operating within the inflammatory pathway of DN. To evaluate the genes associated with the pathogenesis and susceptibility of the disease, we constructed a PPI network to identify hub genes. The analysis of microarray data confirmed that these genes exhibit differential regulation in patients with DN compared to controls. This observation underscores the potential involvement of inflammatory mechanisms in the complex pathophysiology of the disease.
In DN, proteinuria induces renal endothelial injury, triggering the synthesis and production of molecules involved in inflammatory pathways, including the expression of cytokines, chemokines, and adhesion molecules [85]. Cytokines are signaling molecules primarily active in inflammatory processes. These molecules categorize into various classes such as interleukins, tumor necrosis factors, and transforming growth factors (TGFs) [86].
Interleukins are divided into families, and the IL-1 subgroup comprises 11 members: IL-1α, IL-1β, IL-1RA, IL-18, IL-36Ra, IL-36α, IL-37, IL-36β, IL-36γ, IL-38, and IL-33. In this study, three interleukins from this family associated with pathophysiological mechanisms of DN were identified (IL-1α, IL-1β, and IL-1RA). Through meta-analysis, we identified polymorphisms in the IL1β and IL1RA genes potentially associated with increased susceptibility in the development of the complication.
IL-1β is a pro-inflammatory cytokine produced by activated macrophages, playing various roles in biological and pathological cellular functions such as proliferation, differentiation, and apoptosis. IL-1β induces the synthesis of prostaglandins, influx and activation of neutrophils, activation of T cells with cytokine production, and activation of B cells. In addition to promoting the differentiation of pro-inflammatory Th17 cells, it also plays a role in angiogenesis by inducing the production of VEGF-A synergistically with the synthesis of TNF and IL-6 (36; 31). The SNP rs16944 (−511 C/T) identified in this gene is located in the promoter region and involves a cytosine-to-thymine substitution at position −511. This polymorphism has been associated with increased gene transcription and, consequently, an exaggerated synthesis of IL-1β. Therefore, this cytokine polymorphism enhances its pro-inflammatory reactions [31,36].
According to the meta-analysis, patients carrying the T allele exhibited a high susceptibility to ND when compared to homozygotes for the C allele (OR = 1.55; 95% CI = 1.12–1.16; and p = 0.0081). This finding is consistent with studies conducted with Korean [30], Indian [31,34], German [33], Polish [35], and Iraqi [36] patients, demonstrating that individuals with type 2 diabetes mellitus (DM2) carrying this polymorphism have a higher likelihood of developing the complication. The overexpression of IL-1β can induce the proliferation of renal mesangial cells and the formation of extracellular matrix, contributing to renal damage [33,35].
The gene for interleukin-1 receptor antagonist (IL-1RA) encodes the IL-1Ra protein, which acts by inhibiting the activities of IL-1. Thus, it is an anti-inflammatory cytokine that determines the functionality of interleukins IL-1α and IL-1β (37). In our study, the variable number tandem repeat (VNTR) polymorphism corresponding to 86 base pairs in intron 2 was found to be positively associated with susceptibility to ND (OR = 2.42; 95% CI = 1.03–5.65; and p = 0.0420). Our findings align with the studies conducted by Lee et al. [30] and Achyut et al. [31], where the polymorphism was significantly associated with the complication in Indian and Korean patients. According to these studies, the polymorphism reduces the synthesis of this cytokine, and consequently, the amount of IL-1β is altered due to the modulatory activity of IL-1RA on the pro-inflammatory cytokine.
Interleukin-10 (IL-10) is an anti-inflammatory cytokine produced by B lymphocytes, Th2 cells, monocytes, and macrophages. Its anti-inflammatory activity is capable of inhibiting the synthesis of pro-inflammatory cytokines such as IL-1β, IL-1α, IL-6, tumor necrosis factor-alpha (TNF-α), and interferon-gamma (INF-γ) [87]. IL-10 is crucial for the maintenance of renal physiology and is associated with the development of renal lesions, given its predominant anatomical localization in renal mesangial cells. IL-10 promotes the deposition of mesangial immune complexes, contributing to the progression of glomerular injury in ND. The SNP −1082 G/A in the IL10 gene did not show an association with ND susceptibility. Scientific evidence suggests that this polymorphism may attenuate the anti-inflammatory protective effect mediated by the cytokine, thereby intensifying the pro-inflammatory process [88,89].
Another identified anti-inflammatory cytokine is TGF-β1, a growth factor that regulates cell proliferation, differentiation, and growth, while also playing a role in the activation and modulation of the expression of other growth factors, including INF-γ and TNF-α. Additionally, it can promote the differentiation of lineage-specific T helper 17 (Th17) cells or regulatory T cells (Treg). Low concentrations of TGF-β1, in conjunction with cytokines such as IL-6 and IL-21, induce the expression of IL-17 and IL-23 receptors, favoring the differentiation of Th17 cells. In contrast, high concentrations of the factor promote the negative regulation of IL-17 expression, favoring the development of Treg cells [76]. Our study identified two SNPs in the TGF-β1 gene: rs1800470 and rs1800471. Due to the insufficient quantity of studies for SNP rs1800471 (915 G/C), it was not possible to summarize the risk estimate through statistical analysis. However, the meta-analysis conducted for SNP rs1800470 (869 T/C) (OR = 2.14; 95% CI = 0.95–4.82; and p = 0.0669) did not demonstrate an association between the polymorphism and the risk of developing DN. Studies suggest that these polymorphisms can enhance the expression of TGF-β1 in mesangial and endothelial cells of glomeruli, favoring renal hypertrophy and the accumulation of extracellular matrix, which are factors that contribute to the development and progression of the complication [76,90].
TNF-α is a pro-inflammatory cytokine that plays a role in the regulation of cell differentiation, proliferation, and death and is involved in both innate and adaptive immune responses. Studies have demonstrated that the hyperglycemic state in patients with diabetes mellitus induces the production of TNF-α through the negative regulation of CD33 in monocytes. In renal cells, TNF-α is cytotoxic, and its elevation can trigger DN [78]. We identified three SNPs in the TNF-α gene: rs1800629 (−308 G/A), rs1799964 (−1031 T/C), and rs361525 (−238 G/A). It was only possible to conduct a meta-analysis with the SNP −308 G/A, and our statistical analysis showed that patients with the A allele have a higher susceptibility to DN (OR = 1.98; 95% CI = 1.33–2.96; and p = 0.0008). Studies report elevated levels of TNF-α in diabetic patients due to increased gene transcription [34,80,81].
The analysis of the −1031 T/C polymorphism in the TNF-α gene revealed that carriers of the C allele also had a higher risk of developing DN (OR = 2.73; 95% CI = 1.76–4.21; and p = 0.0001). Studies demonstrate that this polymorphism in the promoter region is associated with elevated plasma levels of TNF-α [78].
The most frequently mentioned gene in the literature was CCR5, a receptor for various chemokines such as CCL3, MIP-1-α, CCL4, and MIP-1-β, which stimulates the chemotaxis of T lymphocytes. Studies report that CCR5 is involved in the recruitment of monocytes and their differentiation into macrophages in the glomeruli, playing a significant role in developing glomerulosclerosis and fibrosis in DN [20,21,22,23].
In our meta-analysis, the SNP rs1799987 (59029 G/A), located in the promoter region of the CCR5 gene, showed a higher susceptibility to the development of DN (OR = 1.88; 95% CI = 1.23–2.88; and p = 0.0035). This involves a guanine-to-adenine alteration at position 59029. Studies found report that the presence of the A allele, in homozygosity or heterozygosity, is associated with the development of DN in North American [21] and Japanese DM1 patients [20,22], as well as in Indian [17,23,25] and Polish DM2 patients (OR ranging from 1.39 to 6.17) [24]. Additionally, an increase in the expression of the protein was observed in peripheral blood mononuclear cells of individuals carrying the A allele, suggesting that the polymorphism regulates the expression of the CCR5 gene (17,22–25].
Considering our objective, the pathway enrichment analysis was not performed, since transcriptome analysis was performed to validate the results previously found in the study. Thus, our in silico analyses enabled the identification of three key genes (CCL2, CCL5, and CXCL8) concomitantly found in our systematic review. These genes constitute the core, linking with other genes, and exhibit a higher differential expression in the kidneys of individuals with DN than controls (GSE30529 database) and encode pro-inflammatory cytokines. Therefore, we emphasize these two genes as potential targets that could guide further research and contribute to elucidating the pathogenesis of DN, particularly in association with inflammatory and hemodynamic processes.
The CCL2 gene encodes the homonymous protein, also known as monocyte chemoattractant protein 1 (MCP-1), and is considered one of the key molecules promoting inflammation in DN. Polymorphisms in this gene are associated with an increase in its protein expression and subsequent progressive renal failure. Inflammation in the kidney is predominantly marked by the accumulation of macrophages, which is directly related to the progression of diabetes due to increased hyperglycemia and glycated hemoglobin, favoring renal damage and albuminuria [91].
The macrophages recruited to the renal microenvironment of diabetic patients can promote renal fibrosis. These macrophages stimulate the secretion of IL-1 and TGF-β1. IL-1 can trigger an increase in the pro-inflammatory response, acting as a trigger for other cytokines. The increased expression of TGF-β1, in turn, can induce the production of extracellular matrix, contributing to renal fibrosis. Thus, CCL2/MCP-1 is associated with DN and underlying pathological mechanisms both directly and indirectly [92]. This cytokine is also capable of inducing a fibrotic response in mesangial glomerular cells. This mechanism is mediated by the activation of the NF-Κβ transcription factor in these cells, stimulating the accumulation of macrophages in this region. It is suggested that TGF-β1 stimulates the expression of CCL2/MCP-1, reinforcing the indications of a self-amplifying loop of these cytokines, and promoting the accumulation of extracellular matrix in mesangial cells [93].
CCL5 acts as a potent molecule in signalization for several immune cells, such as monocytes and T cells and its up regulation shows similar effects as CCL2. Sample biopsies of DN patients revealed overexpression of CCL5 in tubular cells, which has a direct association with the magnitude of proteinuria and cell infiltration. Moreover, serum levels of DN patients demonstrated elevated serum levels of this cytokine [94]. Urinary sediment messenger RNAs (mRNAs) of the CCL5 gene have been used as a potential biomarker of kidney disease, especially in DN, due to their significant up-expression [95].
It is well known that several factors involved in the pathogenesis of DM can lead to the activation and accumulation of immune cells in different tissues in patients with diabetes, such as hyperglycemia and the increase of reactive molecules such as [94]. The role of the CXCL8 in DN remains unclear; however, it was previously associated with podocyte damage [96]. Moreover, elevated levels of CXCL8 have been found in the early stages of DN and the level increases with the disease progression and is associated with metabolic disturbances and inflammation. Thus, urinary CXCL8 shows a strong potential as an early predictor of DN [97].
Additionally, our study demonstrates that most SNPs significantly associated with a higher susceptibility to developing DN in our meta-analysis are located in genes encoding pro-inflammatory cytokines. In this context, SNPs in genes for anti-inflammatory cytokines like TGF-β and IL-10 did not pose a risk for DN. A mechanism possibly associated with this is the relationship of the complication with sustained inflammation. Thus, through the obtained data and bioinformatics analyses, we observe an imbalance between the production of anti-inflammatory and pro-inflammatory cytokines, with the latter showing higher expression.
Our study has some limitations. Firstly, it is important to highlight that DM and its complications are complex pathologies involving the interaction of genetic and environmental factors. Some discrepancies found in the meta-analysis results may be explained by differences in sample size, individual genetic background, low statistical power, or challenges in the reproducibility of genetic studies. Additionally, it should be considered that for some SNPs, the meta-analysis was conducted with a considerably low number of studies, which may explain the found heterogeneity [98]. Furthermore, the unavailability of articles and the lack of data in potentially eligible studies led to the exclusion of some articles from our systematic review. Finally, there is a need for studies with larger sample sizes.
Nevertheless, our study identified important biomarkers for DN susceptibility. These biomarkers can assist in clinical practice in the disease. The costs of diagnosing and staging the disease are expensive for health systems [99] and the incorporation of a genetic panel can be a tool in the differential diagnosis and genetic screening of individuals susceptible to complications. In addition, the identification of genetic variants related to the risk of developing DN supports the clinical application of precision medicine, providing personalized health measures to the patient and improving the clinical success [100,101].

5. Conclusions

In conclusion, our study indicated a higher risk of DN development associated with SNPs 59029 G/A, −511 C/T, VNTR 86 bp, −308 G/A, and −1031 T/C. Moreover, the bioinformatics analyses associate the differential expression of CCL2, CCR5 and CXCL8 genes with the etiology and progression of DN due to its intrinsic relationship with inflammatory mechanisms. This study also facilitated the identification of several polymorphisms frequently associated with a higher susceptibility to the development of DN in individuals with diabetes mellitus. These polymorphisms were primarily identified in molecules of a pro-inflammatory nature, and the association of genetic variants with risk factors such as glycemic control, obesity, sedentary lifestyle, hypertension, alcohol consumption, and smoking may increase the risk for the complication. Additionally, bioinformatics analyses enabled the elucidation of differentially expressed hub genes, validating the results found and possibly confirming the central role of these genes in DN. This allows the application of our findings in experimental analyses, given the scarcity of studies on the genes CCL2 and VEGF-A identified in our systematic review, which encode cytokines with potential key roles in DN and its underlying inflammatory mechanisms. Furthermore, the results of this genetic panel allow for the effective application of precision medicine in diabetic patients for a possible prognosis of the complication, contributing to the reduction of expenses for the public health system. Further studies should be suggested at the end of the manuscript for other researchers to follow up and confirm the findings. It is important to emphasize that more studies are needed to confirm and validate our findings aiming at the practical and clinical application of the identified biomarkers.

Supplementary Materials

The following supporting information can be downloaded at:https://www.mdpi.com/article/10.3390/jmp5030024/s1.

Author Contributions

Conceptualization, R.d.S.S. and A.A.d.S.R.; methodology, L.d.S., K.d.F.S., R.d.S.S. and A.A.d.S.R.; software, L.d.S., K.d.F.S., R.d.S.S. and A.A.d.S.R.; validation, R.d.S.S. and A.A.d.S.R.; formal analysis, L.d.S., K.d.F.S., L.d.P.A. and C.C.P.d.C.; investigation, L.d.S., K.d.F.S., C.C.P.d.C., R.d.S.S. and A.A.d.S.R.; resources, R.d.S.S. and A.A.d.S.R.; data curation, L.d.S., K.d.F.S. and C.C.P.d.C., writing—original draft preparation, L.d.S., K.d.F.S., C.C.P.d.C., R.d.S.S. and A.A.d.S.R.; writing—review and editing, L.d.S., K.d.F.S., C.C.P.d.C., R.d.S.S. and A.A.d.S.R.; supervision, R.d.S.S. and A.A.d.S.R.; project administration, A.A.d.S.R.; funding acquisition, A.A.d.S.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Hintsa, S.; Dube, L.; Abay, M.; Angesom, T.; Workicho, A. Determinants of diabetic nephropathy in Ayder Referral Hospital, northern Ethiopia: A case-control study. PLoS ONE 2017, 12, e0173566. [Google Scholar] [CrossRef] [PubMed]
  2. de Lima, R.M.; Dos Anjos, L.R.B.; Alves, T.B.; Coelho, A.S.G.; Pedrino, G.R.; da Silva Santos Cruz, A.H.S.; Reis, A.A.S. Do GST polymorphisms influence in the pathogenesis of diabetic nephropathy? Mol. Cell Endocrinol. 2018, 478, 10–16. [Google Scholar] [CrossRef] [PubMed]
  3. Paul, S.; Ali, A.; Katare, R. Molecular complexities underlying the vascular complications of diabetes mellitus—A comprehensive review. J. Diabetes Complicat. 2020, 34, 107613. [Google Scholar] [CrossRef] [PubMed]
  4. Pichler, R.; Afkarian, M.; Dieter, B.P.; Tuttle, K.R. Immunity and inflammation in diabetic kidney disease: Translating mechanisms to biomarkers and treatment targets. Am. J. Physiol. Ren. Physiol. 2017, 312, F716–F731. [Google Scholar] [CrossRef]
  5. Rayego-Mateos, S.; Morgado-Pascual, J.L.; Opazo-Ríos, L.; Guerrero-Hue, M.; García-Caballero, C.; Vázquez-Carballo, C.; Mas, S.; Sanz, A.B.; Herencia, C.; Mezzano, S.; et al. Pathogenic pathways and therapeutic approaches targeting inflammation in diabetic nephropathy. Int. J. Mol. Sci. 2020, 21, 3798. [Google Scholar] [CrossRef]
  6. Agere, S.A.; Kim, E.Y.; Akhtar, N.; Ahmed, S. Syndecans in chronic inflammatory and autoimmune diseases: Pathological insights and therapeutic opportunities. J. Cell. Physiol. 2018, 233, 6346–6358. [Google Scholar] [CrossRef]
  7. Moreno, J.A.; Moreno, S.; Rubio-Navarro, A.; Gómez-Guerrero, C.; Ortiz, A.; Egido, J. Role of chemokines in proteinuric kidney disorders. Expert. Rev. Mol. Med. 2014, 16, e3. [Google Scholar] [CrossRef]
  8. Persson, F.; Rossing, P. Diagnosis of diabetic kidney disease: State of the art and future perspective. Kidney Int. Suppl. 2018, 8, 2–7. [Google Scholar] [CrossRef]
  9. Xue, R.; Gui, D.; Zheng, L.; Zhai, R.; Wang, F.; Wang, N. Mechanistic insight and management of diabetic nephropathy: Recent progress and future perspective. J. Diabetes Res. 2017, 2017, e1839809. [Google Scholar] [CrossRef]
  10. Cole, J.B.; Florez, J.C. Genetics of diabetes mellitus and diabetes complications. Nat. Rev. Nephrol. 2020, 16, 377–390. [Google Scholar] [CrossRef]
  11. Ouzzani, M.; Hammady, H.; Fedorowicz, Z.; Elmagarmid, A. Rayyan—A web and mobile app for systematic reviews. Syst. Ver. 2016, 5, 1–10. [Google Scholar] [CrossRef] [PubMed]
  12. Egger, M.; Smith, G.D.; Schneider, M.; Minder, C. Bias in meta-analysis detected by a simple, graphical test. BMJ 1997, 315, 629–634. [Google Scholar] [CrossRef]
  13. Egger, M.; Smith, G.D. Meta-analysis: Potentials and promise. BMJ 1997, 315, 1371–1374. [Google Scholar] [CrossRef]
  14. Zhang, D.; Ma, J.; Brismar, K.; Efendic, S.; Gu, H.F. A single nucleotide polymorphism alters the sequence of SP1 binding site in the adiponectin promoter region and is associated with diabetic nephropathy among type 1 diabetic patients in the Genetics of Kidneys in Diabetes Study. J. Diabetes Complicat. 2009, 23, 265–272. [Google Scholar] [CrossRef] [PubMed]
  15. Jaziri, R.; Aubert, R.; Roussel, R.; Emery, N.; Maimaitiming, S.; Bellili, N.; Miot, A.; Saulnier, P.J.; Travert, F.; Hadjadj, S.; et al. Association of ADIPOQ genetic variants and plasma adiponectin isoforms with the risk of incident renal events in type 2 diabetes. Nephrol. Dial. Transpl. 2010, 25, 2231–2237. [Google Scholar] [CrossRef] [PubMed]
  16. Cilenšek, I.; Šeruga, M.; Makuc, J.; Završnik, M.; Petrovič, D. The ALOXA5AP gene (rs38022789) is associated with diabetic nephropathy in Slovenian patients with type 2 diabetes mellitus. Gene 2020, 741, 144551. [Google Scholar] [CrossRef] [PubMed]
  17. Ahluwalia, T.S.; Khullar, M.; Ahuja, M.; Kohli, H.S.; Bhansali, A.; Mohan, V.; Venkatesan, R.; Rai, T.S.; Sud, K.; Singal, P.K. Common variants of inflammatory cytokine genes are associated with risk of nephropathy in type 2 diabetes among Asian Indians. PLoS ONE 2009, 4, e5168. [Google Scholar] [CrossRef]
  18. Moon, J.-Y. Association of polymorphisms in monocyte chemoattractant protein-1 promoter with diabetic kidney failure in Korean patients with type 2 diabetes mellitus. J. Korean Med. Sci. 2007, 22, 810–814. [Google Scholar] [CrossRef]
  19. Raina, P.; Matharoo, K.; Bhanwer, A.J.S. Monocyte chemoattractant protein-1 (MCP-1) g.-2518A4G polymorphism and susceptibility to type 2 diabetes (T2D) and end stage renal disease (ESRD) in the North-West Indian population of Punjab. Ann. Hum. Biol. 2015, 42, 276–282. [Google Scholar] [CrossRef]
  20. Nakajima, K.; Tanaka, Y.; Nomiyama, T.; Ogihara, T.; Ikeda, F.; Kanno, R.; Iwashita, N.; Sakai, K.; Watada, H.; Onuma, T.; et al. RANTES promoter genotype is associated with diabetic nephropathy in type 2 diabetic subjects. Diabetes Care 2003, 26, 892–898. [Google Scholar] [CrossRef]
  21. Mlynarski, W.M.; Placha, G.P.; Wolkow, P.P.; Bochenski, J.P.; Warram, J.H.; Krolewski, A.S. Risk of diabetic nephropathy in type 1 diabetes is associated with functional polymorphisms in RANTES receptor gene (CCR5): A sex-specific effect. Diabetes 2005, 54, 3331–3335. [Google Scholar] [CrossRef] [PubMed]
  22. Mokubo, A.; Tanaka, Y.; Nakajima, K.; Watada, H.; Hirose, T.; Kawasumi, M.; Sakai, K.; Kanazawa, A.; Maeda, S.; Hosokawa, K.; et al. Chemotactic cytokine receptor 5 (CCR5) gene promoter polymorphism (59029A/G) is associated with diabetic nephropathy in Japanese patients with type 2 diabetes: A 10-year longitudinal study. Diabetes Res. Clin. Pract. 2006, 73, 89–94. [Google Scholar] [CrossRef] [PubMed]
  23. Prasad, P.; Tiwari, A.K.; Kumar, K.P.; Ammini, A.C.; Gupta, A.; Gupta, R.; Thelma, B.K. Association of TGFβ1, TNFα, CCR2 and CCR5 gene polymorphisms in type-2 diabetes and renal insufficiency among Asian Indians. BMC Med. Genet. 2007, 8, 20. [Google Scholar] [CrossRef] [PubMed]
  24. Buraczynska, M.; Zukowski, P.; Wacinski, P.; Berger-Smyka, B.; Dragan, M.; Mozul, S. Chemotactic cytokine receptor 5 gene polymorphism: Relevance to microvascular complications in type 2 diabetes. Cytokine 2012, 58, 213–217. [Google Scholar] [CrossRef]
  25. Yadav, A.K.; Kumar, V.; Dutta, P.; Bhansali, A.; Jha, V. Variations in CCR5, but not HFE, ELMO1, or SLC12A3, are associated with susceptibility to kidney disease in north Indian individuals with type 2 diabetes. J. Diabetes 2014, 6, 547–555. [Google Scholar] [CrossRef]
  26. Bessa, S.S.; Hamdy, S.M.; Ali, E.M. Haptoglobin gene polymorphism in type 2 diabetic patients with and without nephropathy: An Egyptian study. Eur. J. Intern. Med. 2007, 18, 489–495. [Google Scholar] [CrossRef]
  27. Conway, B.R.; Savage, D.A.; Brady, H.R.; Maxwell, A.P. Association between haptoglobin gene variants and diabetic nephropathy: Haptoglobin polymorphism in nephropathy susceptibility. Nephron Exp. Nephrol. 2007, 105, 75–79. [Google Scholar] [CrossRef]
  28. Seman, N.A.; Anderstam, B.; Mohamud, W.N.W.; Östenson, C.G.; Brismar, K.; Gu, H.F. Genetic, epigenetic and protein analyses of intercellular adhesion molecule 1 in Malaysian subjects with type 2 diabetes and diabetic nephropathy. J. Diabetes Complicat. 2015, 29, 1234–1239. [Google Scholar] [CrossRef]
  29. Dabhi, B.; Mistry, K.N. Oxidative stress and its association with TNF-α-308 G/C and IL-1α-889 C/T gene polymorphisms in patients with diabetes and diabetic nephropathy. Gene 2015, 562, 197–202. [Google Scholar] [CrossRef]
  30. Lee, S.H.; Ihm, C.-G.; Sohn, S.D.; Lee, T.W.; Kim, M.J.; Koh, G.; Oh, S.J.; Woo, J.-T.; Kim, S.W.; Kim, J.W.; et al. Polymorphisms in interleukin-1ß and interleukin-1 receptor antagonist genes are associated with kidney failure in Korean patients with type 2 diabetes mellitus. Am. J. Nephrol. 2004, 24, 410–414. [Google Scholar] [CrossRef]
  31. Achyut, B.R.; Srivastava, A.; Bhattacharya, S.; Mittal, B. Genetic association of interleukin-1β (-511C/T) and interleukin-1 receptor antagonist (86 bp repeat) polymorphisms with Type 2 diabetes mellitus in North Indians. Clin. Chim. Acta 2007, 377, 163–169. [Google Scholar] [CrossRef] [PubMed]
  32. Szoke, D.; Molnar, B.; Solymosi, N.; Racz, K.; Gergics, P.; Blasko, B.; Vasarhelyi, B.; Vannay, A.; Mandy, Y.; Klausz, G.; et al. Polymorphisms of the ApoE, HSD3B1, IL-1beta and p53 genes are associated with the development of early uremic complications in diabetic patients: Results of a DNA resequencing array study. Int. J. Mol. Med. 2009, 23, 217–227. [Google Scholar]
  33. Stefanidis, I.; Kreuer, K.; Dardiotis, E.; Arampatzis, S.; Eleftheriadis, T.; Hadjigeorgiou, G.M.; Zintzaras, E.; Mertens, P.-R. Association between the interleukin-1b gene (IL1B) C-511T polymorphism and the risk of diabetic nephropathy in type 2 diabetes: A candidate-gene association study. DNA Cell Biol. 2014, 33, 463–468. [Google Scholar] [CrossRef]
  34. Hameed, I.; Masoodi, S.R.; Malik, P.A.; Mir, S.A.; Ghazanfar, K.; Ganai, B.A. Genetic variations in key inflammatory cytokines exacerbate the risk of diabetic nephropathy by influencing the gene expression. Gene 2018, 661, 51–59. [Google Scholar] [CrossRef] [PubMed]
  35. Buraczynska, M.; Ksiazek, K.; Wacinski, P.; Zaluska, W. Interleukin-1β gene (IL1B) polymorphism and risk of developing diabetic nephropathy. Immunol. Investig. 2019, 48, 577–584. [Google Scholar] [CrossRef]
  36. Al-Husseini, R.M.A.H. Impact of interleukin-1 beta gene allelic polymorphisms in diabetic and non-diabetic hemodialysis Iraqi patients. Syst. Rev. Pharm. 2020, 11, 63–69. [Google Scholar]
  37. Blakemore, A.I.F. Interleukin-1 receptor antagonist allele (IL1RN*2) associated with nephropathy in diabetes mellitus. Hum. Genet. 1996, 97, 369–374. [Google Scholar] [CrossRef] [PubMed]
  38. El-Shabrawi, M.M.; Bayoumy, N.M.; Hassan, H.H. Interleukin-4 polymorphism in Egyptian patients with type-2 diabetic nephropathy. Life Sci. J. 2011, 8, 577–582. [Google Scholar]
  39. Neelofar, K.; Ahmad, J.; Ahmad, A.; Alam, K. Study of IL4-590C/T and IL6-174G/C gene polymorphisms in type 2 diabetic patients with chronic kidney disease in North Indian population. J. Cell Biochem. 2017, 118, 1803–1809. [Google Scholar] [CrossRef]
  40. Kitamura, A.; Hasegawa, G.; Obayashi, H.; Kamiuchi, K.; Ishii, M.; Yano, M.; Tanaka, T.; Yamaguchi, M.; Shigeta, H.; Ogata, M.; et al. Interleukin-6 polymorphism (-634C/G) in the promoter region and the progression of diabetic nephropathy in Type 2 diabetes. Diabet. Med. 2002, 19, 1000–1005. [Google Scholar] [CrossRef]
  41. Chang, W.-T.; Huang, M.-C.; Chung, H.-F.; Chiu, Y.-F.; Chen, P.-S.; Chen, F.-P.; Lee, C.-Y.; Shin, S.-J.; Hwang, S.-J.; Huang, Y.-F.; et al. Interleukin-6 gene polymorphisms correlate with the progression of nephropathy in Chinese patients with type 2 diabetes: A prospective cohort study. Diabetes Res. Clin. Pr. 2016, 120, 15–23. [Google Scholar] [CrossRef] [PubMed]
  42. Kitamura, A.; Hasegawa, G.; Obayashi, H.; Kamiuchi, K.; Ishii, M.; Yano, M.; Tanaka, T.; Yamaguchi, M.; Shigeta, H.; Ogata, M.; et al. Association of interleukin-6 polymorphism (-634C/G) in the promoter region with diabetic nephropathy in type 2 diabetes mellitus. Bangladesh J. Med. Sci. 2019, 18, 741–747. [Google Scholar]
  43. Ng, D.P.; Nurbaya, S.; Sandra, H.J.; Krolewski, A.S. An IL-6 haplotype on human chromosome 7p21 confers risk for impaired renal function in type 2 diabetic patients. Kidney Int. 2008, 74, 521–527. [Google Scholar] [CrossRef]
  44. Shanmuganathan, R.; Ramanathan, K.; Padmanabhan, G.; Vijayaraghavan, B. Evaluation of Interleukin 8 gene polymorphism for predicting inflammation in Indian chronic kidney disease and peritoneal dialysis patients. Alex. J. Med. 2016, 53, 215–220. [Google Scholar] [CrossRef]
  45. Rodrigues, K.F.; Pietrani, N.T.; Sandrim, V.C.; Vieira, C.M.A.F.; Fernandes, A.P.; Bosco, A.A.; Gomes, K.B. Association of a large panel of cytokine gene polymorphisms with complications and comorbidities in type 2 diabetes patients. J. Diabetes Res. 2015, 2015, 605965. [Google Scholar] [CrossRef] [PubMed]
  46. Yin, Q.; Zhai, Q.; Wang, D.; Hai, J.; Cao, M.; Wang, J.; Wang, T. Investigation on the association between interleukin-10 -592C/A, 819C/T and -1082A/G gene polymorphisms and development of diabetic nephropathy. Int. J. Clin. Exp. Pathol. 2015, 8, 15216–15221. [Google Scholar]
  47. Ma, D.H.; Xu, Q.Y.; Liu, Y.; Zhai, Q.Q.; Guo, M.H. Association between interleukin-10 gene polymorphisms and susceptibility to diabetic nephropathy in a Chinese population. Genet. Mol. Res. 2016, 15, 1–7. [Google Scholar] [CrossRef]
  48. Polina, E.R.; da Silva Pereira, B.L.; Crispim, D.; Sbruzzi, R.C.; Canani, L.H.; Dos Santos, K.G. Association of -1082A>G polymorphism in the interleukin-10 gene with estimated glomerular filtration rate in type 2 diabetes. Kidney Blood Press. Res. 2017, 42, 1164–1174. [Google Scholar] [CrossRef]
  49. Fathy, S.A.; Mohamed, M.R.; Ali, M.A.; El-Helaly, A.E.; Alattar, A.T. Influence of IL-6, IL-10, IFN-γ, and TNF-α genetic variants on susceptibility to diabetic kidney disease in type 2 diabetes mellitus patients. Biomarkers 2019, 24, 43–55. [Google Scholar] [CrossRef]
  50. Kung, W.J.; Lin, C.C.; Liu, S.H.; Chaung, H.C. Association of interleukin-10 polymorphisms with cytokines in type 2 diabetic nephropathy. Diabetes Technol. Ther. 2010, 12, 809–813. [Google Scholar] [CrossRef]
  51. Liao, L.-N.; Chen, C.-C.; Wu, F.-Y.; Lin, C.-C.; Hsiao, J.-H.; Chang, C.-T.; Kardia, S.L.R.; Li, T.-C.; Tsai, F.-J. Identified single-nucleotide polymorphisms and haplotypes at 16q22.1 increase diabetic nephropathy risk in Han Chinese population. BMC Genet. 2014, 15, 113. [Google Scholar] [CrossRef] [PubMed]
  52. Wang, Y.; Luk, A.O.Y.; Ma, R.C.W.; So, W.-Y.; Tam, C.H.T.; Ng, M.C.Y.; Yang, X.; Lam, V.; Tong, P.C.Y.; Chan, J.C.N. Predictive role of multilocus genetic polymorphisms in cardiovascular disease and inflammation-related genes on chronic kidney disease in Type 2 diabetes—An 8-year prospective cohort analysis of 1163 patients. Nephrol Dial Transpl. 2012, 27, 190–196. [Google Scholar] [CrossRef] [PubMed]
  53. Albert, C.; Kube, J.; Albert, A.; Schanze, D.; Zenker, M.; Mertens, P.R. Cubilin single nucleotide polymorphism variants are associated with macroangiopathy while a matrix metalloproteinase-9 single nucleotide polymorphism flip-flop may indicate susceptibility of diabetic nephropathy in type-2 diabetic patients. Nephron 2019, 141, 156–165. [Google Scholar] [CrossRef] [PubMed]
  54. Buraczynska, K.; Koziol-Montewka, M.; Majdan, M.; Tokarz, A.; Ksiazek, A. Genetic determination of TNF and myeloperoxidase production in dialyzed patients with diabetic nephropathy. Ren. Fail. 2004, 26, 633–639. [Google Scholar] [CrossRef]
  55. Katakami, N.; Kume, S.; Kaneto, H.; Uzu, T.; Kashiwagi, A.; Yamasaki, Y.; Maegawa, H.; Shimomura, I. Association of myeloperoxidase G-463A gene polymorphism with diabetic nephropathy in Japanese type 2 diabetic subjects. Endocr. J. 2013, 60, 457–471. [Google Scholar] [CrossRef]
  56. Gautam, A.; Gupta, S.; Mehndiratta, M.; Sharma, M.; Singh, K.; Kalra, O.P.; Agarwal, S.; Gambhir, J.K. Association of NFKB1 gene polymorphism (rs28362491) with levels of inflammatory biomarkers and susceptibility to diabetic nephropathy in Asian Indians. World J. Diabetes 2017, 8, 66–73. [Google Scholar] [CrossRef]
  57. Chen, F.; Li, Y.M.; Yang, L.Q.; Zhong, C.G.; Zhuang, Z.X. Association of NOS2 and NOS3 gene polymorphisms with susceptibility to type 2 diabetes mellitus and diabetic nephropathy in the Chinese Han population. IUBMB Life 2016, 68, 516–525. [Google Scholar] [CrossRef]
  58. Cheema, B.S.; Iyengar, S.; Ahluwalia, T.S.; Kohli, H.S.; Sharma, R.; Shah, V.N.; Bhansali, A.; Sakhuja, V.; Khullar, M. Association of an osteopontin gene promoter polymorphism with susceptibility to diabetic nephropathy in Asian Indians. Clin. Chim. Acta 2012, 413, 1600–1604. [Google Scholar] [CrossRef]
  59. Liu, L.; Zheng, T.; Wang, F.; Wang, N.; Song, Y.; Li, M.; Li, L.; Jiang, J.; Zhao, W. Pro12Ala polymorphism in the PPARG gene contributes to the development of diabetic nephropathy in Chinese type 2 diabetic patients. Diabetes Care 2010, 33, 144–149. [Google Scholar] [CrossRef]
  60. Li, C.; Lei, T. Rs12976445 polymorphism is associated with risk of diabetic nephropathy through modulating expression of MicroRNA-125 and Interleukin-6R. Med. Sci. Monit. 2015, 21, 3490–3497. [Google Scholar] [CrossRef]
  61. Araki, S.-I.; Ng, D.P.K.; Krolewski, B.; Wyrwicz, L.; Rogus, J.J.; Canani, L.; Makita, Y.; Haneda, M.; Warram, J.H.; Krolewski, A.S. Identification of a common risk haplotype for diabetic nephropathy at the Protein Kinase C-β1 (PRKCB1) gene locus. J. Am. Soc. Nephrol. 2003, 14, 2015–2024. [Google Scholar] [CrossRef]
  62. Zhu, H.; Yu, W.; Xie, Y.; Zhang, H.; Bi, Y.; Zhu, D. Association of Pentraxin 3 gene polymorphisms with susceptibility to diabetic nephropathy. Med. Sci. Monit. 2017, 23, 428–436. [Google Scholar] [CrossRef] [PubMed]
  63. El Naidany, S.S.; Zahran, E.; Abd El Gayed, E.M. Association of Pentraxin 3 rs2305619 (A/G) gene polymorphism and its serum level with the risk of nephropathy in type II diabetic patients. Gene Rep. 2020, 20, 100670. [Google Scholar] [CrossRef]
  64. Lindholm, E.; Bakhtadze, E.; Sjögren, M.; Cilio, C.M.; Agardh, E.; Groop, L.; Agardh, C.D. The -374 T/A polymorphism in the gene encoding RAGE is associated with diabetic nephropathy and retinopathy in type 1 diabetic patients. Diabetologia 2006, 49, 2745–2755. [Google Scholar] [CrossRef] [PubMed]
  65. Cai, W.; Li, J.; Xu, J.-X.; Liu, Y.; Zhang, W.; Xiao, J.-R.; Zhu, L.-Y.; Liu, J.-Y. Association of 2184AG polymorphism in the RAGE gene with diabetic nephropathy in Chinese patients with type 2 diabetes. J. Diabetes Res. 2015, 2015. [Google Scholar] [CrossRef]
  66. McDonough, C.W.; Palmer, N.D.; Hicks, P.J.; Roh, B.H.; An, S.S.; Cooke, J.N.; Hester, J.M.; Wing, M.R.; Bostrom, M.A.; Rudock, M.E.; et al. A genome-wide association study for diabetic nephropathy genes in African Americans. Kidney Int. 2011, 79, 563–572. [Google Scholar] [CrossRef]
  67. Li, F.; Mao, A.; Fu, X.; She, Y.; Wei, X. Correlation between SEPS1 gene polymorphism and type 2 diabetes mellitus: A preliminary study. J. Clin. Lab. Anal. 2019, 33, e22967. [Google Scholar] [CrossRef]
  68. Bodhini, D.; Chidambaram, M.; Liju, S.; Revathi, B.; Laasya, D.; Sathish, N.; Kanthimathi, S.; Ghosh, S.; Anjana, R.M.; Mohan, V.; et al. Association of rs11643718 SLC12A3 and rs741301 ELMO1 variants with diabetic nephropathy in South Indian population. Ann. Hum. Genet. 2016, 80, 336–341. [Google Scholar] [CrossRef]
  69. Sinha, N.; Yadav, A.K.; Kumar, V.; Dutta, P.; Bhansali, A.; Jha, V. SUMO4 163 G>A variation is associated with kidney disease in Indian subjects with type 2 diabetes. Mol. Biol. Rep. 2016, 43, 345–348. [Google Scholar] [CrossRef]
  70. Patel, A.; Hibberd, M.L.; Millward, B.A.; Demaine, A.G. Chromosome 7q35 and susceptibility to diabetic microvascular complications. J. Diabetes Complicat. 1996, 10, 62–67. [Google Scholar] [CrossRef]
  71. Wong, T.Y.H.; Poon, P.; Chow, K.M.; Szeto, C.C.; Cheung, M.K.; Li, P.K.T. Association of transforming growth factor-beta (TGF-β) T869C (Leu 10Pro) gene polymorphisms with type 2 diabetic nephropathy in Chinese. Kidney Int. 2003, 63, 1831–1835. [Google Scholar] [CrossRef]
  72. Patel, A.; Scott, W.R.; Lympany, P.A.; Rippin, J.D.; Gill, G.V.; Barnett, A.H.; Bain, S.C.; Warren 3/UK GoKind Study Group. The TGF-β1 gene codon 10 polymorphism contributes to the genetic predisposition to nephropathy in Type 1 diabetes. Diabet. Med. 2005, 22, 69–73. [Google Scholar] [CrossRef]
  73. Buraczynska, M.; Baranowicz-Gaszczyk, I.; Borowicz, E.; Ksiazek, A. TGF-β1 and TSC-22 gene polymorphisms and susceptibility to microvascular complications in type 2 diabetes. Nephron Physiol. 2007, 106, 69–75. [Google Scholar] [CrossRef]
  74. Valladares-Salgado, A.; Angeles-Martínez, J.; Rosas, M.; García-Mena, J.; Utrera-Barillas, D.; Gómez-Díaz, R.; Escobedo-de la Peña, J.; Parra, E.J.; Cruz, M. Association of polymorphisms within the transforming growth factor-b1 gene with diabetic nephropathy and serum cholesterol and triglyceride concentrations. Nephrology 2010, 15, 644–648. [Google Scholar] [CrossRef] [PubMed]
  75. Mou, X.; Liu, W.-h.; Zhou, D.-y.; Liu, Y.-h.; Hu, Y.-b.; Ma, G.-l.; Shou, C.-m.; Chen, J.-w.; Zhao, J.-x. Association of Chinese Medicine Constitution Susceptibility to Diabetic Nephropathy and Transforming Growth Factor-β1 (T869C) Gene Polymorphism. Chin. J. Integr. Med. 2011, 17, 680–684. [Google Scholar] [CrossRef] [PubMed]
  76. El-Sherbini, S.M.; Shahen, S.M.; Mosaad, Y.M.; Abdelgawad, M.S.; Talaat, R.M. Gene polymorphism of transforming growth factor-b1 in Egyptian patients with type 2 diabetes and diabetic nephropathy. Acta Biochim. Biophys. Sin. 2013, 45, 330–338. [Google Scholar] [CrossRef] [PubMed]
  77. Abbas, S.A.; Raza, S.T.; Mir, S.S.; Siddiqi, Z.; Zaidi, A.; Zaidi, Z.H.; Mahdi, F. Role of variants rs5030717 and rs5030718 of TLR4 in the risk prediction of nephropathy, hypertension and dyslipidemia in type 2 diabetes mellitus. Br. J. Biomed. Sci. 2018, 75, 163–168. [Google Scholar] [CrossRef] [PubMed]
  78. Gupta, S.; Mehndiratta, M.; Kalra, S.; Kalra, O.P.; Shukla, R.; Gambhir, J.K. Association of Tumor Necrosis Factor (TNF) promoter polymorphisms with plasma TNF-α levels and susceptibility to diabetic nephropathy in North Indian population. J. Diabetes Complicat. 2015, 29, 338–342. [Google Scholar] [CrossRef]
  79. Emara, M.; El-Edel, R.; Fathy, W.M.; Aboelkhair, N.T.; Watany, M.M.; Abou-Elela, D.H. Study the Association of Tumor Necrosis Factor Promoter Polymorphism with Type 2 Diabetic Nephropathy. Mediat. Inflamm. 2020, 2020, 1498278. [Google Scholar] [CrossRef]
  80. Peng, Y.; Li, L.J. TNF-α-308G/A polymorphism associated with TNF-α protein expression in patients with diabetic nephropathy. Int. J. Clin. Exp. Pathol. 2015, 8, 3127–3131. [Google Scholar]
  81. Umapathy, D.; Krishnamoorthy, E.; Mariappanadar, V.; Viswanathan, V.; Ramkumar, K.M. Increased levels of circulating TNF-α is associated with (-308G/A) promoter polymorphism of TNF-α gene in Diabetic Nephropathy. Int. J. Biol. Macromol. 2018, 107, 2113–2121. [Google Scholar] [CrossRef] [PubMed]
  82. Kumar, V.; Yadav, A.K.; Kumar, V.; Bhansali, A.; Jha, V. Uromodulin rs4293393 T>C variation is associated with kidney disease in patients with type 2 diabetes. Indian. J. Med. Res. 2017, 146, 15–21. [Google Scholar]
  83. Yang, B.; Cross, D.F.; Ollerenshaw, M.; Millward, B.A.; Demaine, A.G. Polymorphisms of the vascular endothelial growth factor and susceptibility to diabetic microvascular complications in patients with type 1 diabetes mellitus. J. Diabetes Complicat. 2003, 17, 1–6. [Google Scholar] [CrossRef] [PubMed]
  84. Matoba, K.; Takeda, Y.; Nagai, Y.; Kawanami, D.; Utsunomiya, K.; Nishimura, R. Unraveling the Role of Inflammation in the Pathogenesis of Diabetic Kidney Disease. Int. J. Mol. Sci. 2019, 20, 3393. [Google Scholar] [CrossRef] [PubMed]
  85. Zandi-Nejad, K.; Eddy, A.A.; Glassock, R.J.; Brenner, B.M. Why is proteinuria an ominous biomarker of progressive kidney disease? Kidney Int. 2004, 66, S76–S89. [Google Scholar] [CrossRef]
  86. Kany, S.; Vollrath, J.T.; Relja, B. Cytokines in inflammatory disease. Int. J. Mol. Sci. 2019, 20, 6008. [Google Scholar] [CrossRef]
  87. Myśliwska, J.; Zorena, K.; Semetkowska-Jurkiewicz, E.; Rachoń, D.; Suchanek, H.; Myśliwski, A. High levels of circulating interleukin-10 in diabetic nephropathy patients. Eur. Cytokine Netw. 2005, 16, 117–122. [Google Scholar]
  88. Rodrigues, K.F.; Pietrani, N.T.; Bosco, A.A.; Campos, F.M.F.; Sandrim, V.C.; Gomes, K.B. IL-6, TNF-α, and IL-10 levels/polymorphisms and their association with type 2 diabetes mellitus and obesity in Brazilian individuals. Arch. Endocrinol. Metab. 2017, 61, 438–446. [Google Scholar] [CrossRef]
  89. Yin, Y.-W.; Hu, A.-M.; Sun, Q.-Q.; Zhang, B.-B.; Liu, H.-L.; Wang, Q.; Zeng, Y.-H.; Xu, R.-J.; Zhang, S.-J.; Shi, L.-B. Association between interleukin 10 gene -1082 A/G polymorphism and the risk of type 2 diabetes mellitus: A meta-analysis of 4250 subjects. Cytokine 2013, 62, 226–231. [Google Scholar] [CrossRef]
  90. Ziyadeh, F.N. Different roles for TGF-beta and VEGF in the pathogenesis of the cardinal features of diabetic nephropathy. Diabetes Res. Clin. Pr. 2008, 82, S38–S41. [Google Scholar] [CrossRef]
  91. Tesch, G.H. MCP-1/CCL2, A new diagnostic marker and therapeutic target for progressive renal injury in diabetic nephropathy. Am. J. Physiol. Ren. Physiol. 2008, 294, F697–F701. [Google Scholar] [CrossRef] [PubMed]
  92. El Mesallamy, H.O.; Ahmed, H.H.; Bassyouni, A.A.; Ahmed, A.S. Clinical significance of inflammatory and fibrogenic cytokines in diabetic nephropathy. Clin. Biochem. 2012, 45, 646–650. [Google Scholar] [CrossRef] [PubMed]
  93. Mezzano, S.; Aros, C.; Droguett, A.; Burgos, M.E.; Ardiles, L.; Flores, C.; Schneider, H.; Ruiz-Ortega, M.; Egido, J. NF-κB activation and overexpression of regulated genes in human diabetic nephropathy. Nephrol. Dial. Transplant. 2004, 19, 2505–2512. [Google Scholar] [CrossRef] [PubMed]
  94. Navarro-González, J.F.; Mora-Fernández, C.; Fuentes, M.M.; García-Pérez, J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat. Rev. Nephrol. 2011, 7, 327–340. [Google Scholar] [CrossRef]
  95. Feng, S.T.; Yang, Y.; Yang, J.F.; Gao, Y.M.; Cao, J.Y.; Li, Z.L.; Tang, T.T.; Lv, L.L.; Wang, B.; Wen, Y.; et al. Urinary sediment CCL5 messenger RNA as a potential prognostic biomarker of diabetic nephropathy. Clin. Kidney J. 2021, 15, 534–544. [Google Scholar] [CrossRef]
  96. Barutta, F.; Bellini, S.; Gruden, G. Mechanisms of podocyte injury and implications for diabetic nephropathy. Clin. Sci. 2022, 136, 493–520. [Google Scholar] [CrossRef]
  97. He, Y.; Li, H.; Wang, R.; Ma, N.; Liu, L.; Shi, R.; Zhang, B.; Lin, N.; Tian, Y. Potential role and expression level of urinary CXCL8 in different stages of incipient diabetic nephropathy with undiminished creatinine clearance: A pilot study. Diabetes Metab. Syndr. Obes. 2023, 16, 1783–1790. [Google Scholar] [CrossRef]
  98. Higgins, J.P.T.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring inconsistency in meta-analyses. BMJ. 2003, 327, 557–560. [Google Scholar] [CrossRef]
  99. Zhou, Z.; Paresh, C.; Hongbo, Y.; Anna, P.F.; Jing, Z.; Ernest, H.L.; Eric, Q.W.; Ruixuan, J.; Raafat, S. Healthcare resource use, costs, and disease progression associated with diabetic nephropathy in adults with type 2 diabetes: A retrospective observational study. Diabetes Ther. 2017, 8, 555–571. [Google Scholar] [CrossRef]
  100. Giuseppe, N.; Cinzia, C.; Paola, B.; Marisa, P.A.; Eric, A. Genetic tests and genomic biomarkers: Regulation, qualification and validation. Clin. Cases Min. Bone Metab. 2008, 5, 149–154. [Google Scholar]
  101. Matthew, J.S.; Maheswari, S.; Nathan, R.W. Making Meaningful Clinical Use of Biomarkers. Biomark. Insights 2017, 12, 1177271917715236. [Google Scholar]
Figure 1. PRISMA flowchart detailing the exclusion and inclusion process of studies in the systematic review.
Figure 1. PRISMA flowchart detailing the exclusion and inclusion process of studies in the systematic review.
Jmp 05 00024 g001
Figure 2. Graphic for the methodological quality analysis of: (A) case-control studies; (B) cohort studies; and (C) cross-sectional studies.
Figure 2. Graphic for the methodological quality analysis of: (A) case-control studies; (B) cohort studies; and (C) cross-sectional studies.
Jmp 05 00024 g002
Figure 3. Forest plot for genotypic comparison of SNP 59029 G/A in the CCR5 gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Figure 3. Forest plot for genotypic comparison of SNP 59029 G/A in the CCR5 gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Jmp 05 00024 g003
Figure 4. Forest plot for genotypic comparison of SNP −511 C/T in IL1B gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Figure 4. Forest plot for genotypic comparison of SNP −511 C/T in IL1B gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Jmp 05 00024 g004
Figure 5. Forest plot for genotypic comparison of SNP 869 T/C in TGF-1B gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Figure 5. Forest plot for genotypic comparison of SNP 869 T/C in TGF-1B gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Jmp 05 00024 g005
Figure 6. Forest plot for genotypic comparison of SNP −1082 G/A in IL10 gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Figure 6. Forest plot for genotypic comparison of SNP −1082 G/A in IL10 gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Jmp 05 00024 g006
Figure 7. Forest plot for genotypic comparison of the variable number tandem repeat (VNTR) polymorphism of 86 bp in IL-1RN gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Figure 7. Forest plot for genotypic comparison of the variable number tandem repeat (VNTR) polymorphism of 86 bp in IL-1RN gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Jmp 05 00024 g007
Figure 8. Forest plot for genotypic comparison of SNP −308 G/A in TNF gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Figure 8. Forest plot for genotypic comparison of SNP −308 G/A in TNF gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Jmp 05 00024 g008
Figure 9. Forest plot for genotypic comparison of SNP −1031 T/C in TNF gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Figure 9. Forest plot for genotypic comparison of SNP −1031 T/C in TNF gene. The odds ratio (OR) and 95% confidence interval (95% CI) were calculated using the Mantel–Haenszel test (random-effects model) due to the value obtained from the heterogeneity test (I2).
Jmp 05 00024 g009
Figure 10. Funnel plot for publication bias analysis of studies for SNPs. (A): 59029 G/A; (B): −511 C/T; (C): 869 T/C; (D): −1082 G/A; (E): VNTR of 86 bp; (F): −308 G/A; and (G): −1031 T/C.
Figure 10. Funnel plot for publication bias analysis of studies for SNPs. (A): 59029 G/A; (B): −511 C/T; (C): 869 T/C; (D): −1082 G/A; (E): VNTR of 86 bp; (F): −308 G/A; and (G): −1031 T/C.
Jmp 05 00024 g010
Figure 11. STRING protein network with 34 Nodes and 261 edges associated with the inflammatory pathway.
Figure 11. STRING protein network with 34 Nodes and 261 edges associated with the inflammatory pathway.
Jmp 05 00024 g011
Figure 12. Protein–protein interaction (PPI) network with the 20 centralized hub genes.
Figure 12. Protein–protein interaction (PPI) network with the 20 centralized hub genes.
Jmp 05 00024 g012
Figure 13. (A) Transcriptomic analysis workflow of control and DN groups. (B) Volcano plot representing differentially expressed genes found in the transcriptomic analysis of control and DN groups considering a fold change of 1.2. Blue: down-regulated genes; red: up-regulated genes; and gray: non-significant genes.
Figure 13. (A) Transcriptomic analysis workflow of control and DN groups. (B) Volcano plot representing differentially expressed genes found in the transcriptomic analysis of control and DN groups considering a fold change of 1.2. Blue: down-regulated genes; red: up-regulated genes; and gray: non-significant genes.
Jmp 05 00024 g013
Table 1. PEO acronym with respective descriptions.
Table 1. PEO acronym with respective descriptions.
AcronymDescription
P (Population)Individuals with diabetes mellitus (types 1 and 2) who developed Diabetic Nephropathy as a complication.
E (ExposurePresence of genetic polymorphisms associated with inflammation in the population.
O (Outcome)Outcome of interest related to the development of Diabetic Nephropathy.
Table 2. Search strategies developed for systematic review.
Table 2. Search strategies developed for systematic review.
DatabasesSearch Strategy
Pubmed(((‘Diabetic Nephropathies’) OR (Nephropathies, Diabetic) OR (Nephropathy, Diabetic) OR (‘Diabetic Nephropathy’) OR (‘Diabetic Kidney Disease’) OR (‘Diabetic Kidney Diseases’) OR (Kidney Disease *, Diabetic) OR (‘Diabetic Glomerulosclerosis’) OR (Glomerulosclerosis, Diabetic) OR (‘Intracapillary Glomerulosclerosis’) OR (‘Nodular Glomerulosclerosis’) OR (Glomerulosclerosis, Nodular) OR (‘Kimmelstiel-Wilson Syndrome’) OR (‘Kimmelstiel Wilson Syndrome’) OR (Syndrome, Kimmelstiel-Wilson) OR (‘Kimmelstiel-Wilson Disease’) OR (‘Kimmelstiel Wilson Disease’)) AND ((Polymorphism *, Genetic) OR (‘Genetic Polymorphism’) OR (‘Genetic Polymorphisms’) OR (‘Gene Polymorphism’) OR (‘Gene Polymorphisms’) OR (Polymorphism *, Gene) OR (Polymorphism * (Genetics)) OR (‘Genetic Susceptibility’) OR (‘Genetic Susceptibilities’) OR (Susceptibilit *, Genetic) OR (‘Genetic Predisposition’) OR (‘Genetic Predispositions’) OR (Predisposition *, Genetic)) AND ((Inflammation *) OR (‘Innate Inflammatory Response’) OR (‘Innate Inflammatory Responses’) OR (Inflammatory Response, Innate) OR (‘Inflammation Mediators’) OR (Mediators, Inflammation) OR (‘Mediators of Inflammation’)))
SCOPUS(TITLE-ABS-KEY (“Diabetic Nephropath *”) OR TITLE-ABS-KEY (Nephropath *, Diabetic) OR TITLE-ABS-KEY (“Diabetic Kidney Disease *”) OR TITLE-ABS-KEY (Kidney Disease *, Diabetic) OR TITLE-ABS-KEY (“Diabetic Glomerulosclerosis”) OR TITLE-ABS-KEY (Glomerulosclerosis, Diabetic) OR-TITLE-ABS-KEY (“Intracapillary Glomerulosclerosis”) OR TITLE-ABS-KEY (“Nodular Glomerulosclerosis”) OR TITLE-ABS-KEY (Glomerulosclerosis, Nodular) OR TITLE-ABS-KEY (“Kimelstiel$Wilson Syndrome”) OR TITLE-ABS-KEY (Syndrome, Kimmelstiel-Wilson) OR TITLE-ABS-KEY (“Kimmelstiel$Wilson Disease”) AND TITLE-ABS-KEY (Polymorphism *, Genetic) OR TITLE-ABS-KEY (“Genetic Polymorphism *”) OR TITLE-ABS-KEY (“Gene Polymorphism *”) OR TITLE-ABS-KEY (“Polymorphism, Gene”) OR TITLE-ABS-KEY (“Polymorphism (Genetics)”) OR TITLE-ABS-KEY (“Genetic Susceptibilit *”) OR TITLE-ABS-KEY (Susceptibilit *, Genetic) OR TITLE-ABS-KEY (“Genetic Predisposition *”) OR TITLE-ABS-KEY (“Predisposition, Genetic”) AND TITLE-ABS-KEY (Inflammat *) OR TITLE-ABS-KEY (“Innate Inflammatory Response”) OR TITLE-ABS-KEY (Inflammatory Response, Innate) OR TITLE-ABS-KEY (“Inflammation Mediators”) OR TITLE-ABS-KEY (Mediators, Inflammation) OR TITLE-ABS-KEY (“Mediators of Inflammation”))
BVS(‘Diabetic Nephropathies’) OR (Nephropath *, Diabetic) OR (‘Diabetic Nephropathy’) OR (‘Diabetic Kidney Disease’) OR (‘Diabetic Kidney Diseases’) OR (Kidney Disease *, Diabetic) OR (‘Diabetic Glomerulosclerosis’) OR (Glomerulosclerosis, Diabetic) OR (‘Intracapillary Glomerulosclerosis’) OR (‘Nodular Glomerulosclerosis’) OR (Glomerulosclerosis, Nodular) OR (‘Kimmelstiel-Wilson Syndrome’) OR (‘Kimmelstiel Wilson Syndrome’) OR (Syndrome, Kimmelstiel-Wilson) OR (‘Kimmelstiel- Wilson Disease’) OR (‘Kimmelstiel Wilson Disease’) AND (Polymorphism *, Genetic) OR (‘Genetic Polymorphism’) OR (‘Genetic Polymorphisms’) OR (‘Gene Polymorphism’) OR (‘Gene Polymorphisms’) OR (Polymorphism *, Gene) OR (Polymorphism * (Genetics)) OR (‘Genetic Susceptibility’) OR (‘Genetic Susceptibilities’) OR (Susceptibilit *, Genetic) OR (‘Genetic Predisposition’) OR (‘Genetic Predispositions’) OR (Predisposition *, Genetic) AND (Inflammat *) OR (‘Innate Inflammatory Response’) OR (Inflammatory Response, Innate) OR (‘Innate Inflammatory Responses’) OR (‘Inflammation Mediators’) OR (Mediators, Inflammation) OR (‘Mediators of Inflammation’)
Web of ScienceTS = (((“Diabetic Nephropathies”) OR (Nephropathies, Diabetic) OR (Nephropathy, Diabetic) OR (“Diabetic Nephropathy”) OR (“Diabetic Kidney Disease$”) OR (Kidney Disease$, Diabetic) OR (“Diabetic Glomerulosclerosis”) OR (Glomerulosclerosis, Diabetic) OR (“Intracapillary Glomerulosclerosis”) OR (“Nodular Glomerulosclerosis”) OR (Glomerulosclerosis, Nodular) OR (“Kimmelstiel-Wilson Syndrome”) OR (“Kimmelstiel Wilson Syndrome”) OR (Syndrome, Kimmelstiel-Wilson) OR (“Kimmelstiel-Wilson Disease”) OR (“Kimmelstiel Wilson Disease”)) AND ((Polymorphism *, Genetic) OR (“Genetic Polymorphism$”) OR (“Gene Polymorphism$”) OR (Polymorphism$, Gene) OR (Polymorphism$ (Genetics)) OR (“Genetic Susceptibilit *”) OR (Susceptibilit *, Genetic) OR (“Genetic Predisposition$”) OR (Predisposition$, Genetic)) AND ((Inflammat *) OR (“Innate Inflammatory Response$”) OR (Inflammatory Response, Innate) OR (“Inflammation Mediators”) OR (Mediators, Inflammation) OR (“Mediators of Inflammation”)))
EMBASE(‘diabetic glomerulopathy’ OR ‘diabetic glomerulosclerosis’ OR ‘diabetic intercapillary glomerulosclerosis’ OR ‘diabetic kidney disease’ OR ‘diabetic nephropathies’ OR ‘glomerulonecrosis, intercapillary’ OR ‘glomerulosclerosis, diabetic’ OR ‘glomerulosclerosis, intercapillary’ OR ‘intercapillary glomerulosclerosis’ OR ‘diabetic nephropathy’ OR ‘kimmelstiel wilson nephropathy’ OR ‘kimmelstiel wilson syndrome’ OR ‘nephropathy, diabetic’ OR ‘kidney disease’ OR ‘diabetic complication’) AND (‘genetic polymorphism’ OR ‘polymorphism (genetics)’ OR ‘polymorphism, genetic’ OR ‘genetic susceptibility’ OR ‘genetic predisposition’) AND (inflammation OR ‘acute inflammation’ OR ‘inflammation reaction’ OR ‘inflammation response’ OR ‘inflammatory condition’ OR ‘inflammatory lesion’ OR ‘inflammatory process’ OR ‘inflammatory reaction’ OR ‘inflammatory response’ OR ‘reaction, inflammation’ OR ‘response, inflammatory’ OR ‘innate immunity’ OR ‘autacoid’ OR ‘inflammation mediators’)
This term (*) promotes an amplification to results of the search. $ promotes an amplification to the search to association, for example disease and diseases.
Table 3. Genes and polymorphisms associated with the inflammatory pathway of DN.
Table 3. Genes and polymorphisms associated with the inflammatory pathway of DN.
GeneGene LocationGene Function in the Inflammatory PathwayEvaluated PolymorphismType of PolymorphismReferences
ADIPOQ3q27.3It encodes an anti-inflammatory adipocytokine that antagonizes TNF-alpha by negatively regulating its expression in various tissues and organs and neutralizing its effects. It prevents NF-kappa-B endothelial signaling through a cAMP-dependent pathway.rs266729
(−11377 C/G)
SNP[14]
rs17300539 (−11391 G/A)[15]
rs2241766
(45 T/G)
ALOXA5AP13q12.3It encodes a protein, that when associated with 5-lipoxygenase, performs the synthesis of leukotrienes (LTs). Leukotrienes are a family of lipid mediators that act as pro-inflammatory mediators.rs3803278
(8733 T/C)
SNP[16]
CCL2 (MCP-1)17q12Encodes a chemokine member of the CC subfamily involved in immunoregulatory and inflammatory processes. Displays chemotactic activity for monocytes and basophils, but not for neutrophils or eosinophils.rs3917887 (int1del554-567)Indel[17]
rs1024611

(−2518 A/G)
SNP[18,19]
CCR53p21.31Chemokine CC receptors (CCRs) predominantly recognize inflammatory CC chemokines, such as CCL3, CCL4, and RANTES. They may play a role in controlling the proliferation or differentiation of the granulocytic lineage and participate in the migration of T lymphocytes to the site of infection, acting as a chemotactic receptor.rs1799987
(59029 G/A)
SNP[17,20,21,22,23,24,25]
rs333
(Delta 32)
Indel[17,21]
Hp16q22.2Haptoglobin (Hp) captures hemoglobin and combines it with free plasma, enabling hepatic recycling of heme iron and preventing renal damage. It also acts as an antioxidant, antibacterial agent, and plays a role in modulating the acute-phase inflammatory response.Hp1/Hp2Codominant alleles[26,27]
ICAM119p13.2Encodes a cell surface glycoprotein that is particularly expressed in endothelial cells and the immune system. It binds to integrins of the CD11a/CD18 or CD11b/CD18 type.rs5498 (K469E; 1462 A/G)SNP[28]
IL1α2q14.1Encodes a member of the interleukin-1 cytokine family. It is pleiotropic and related to various immune responses, inflammatory processes, and hematopoiesis. This cytokine is produced by monocytes and macrophages as a pro-protein, which is proteolytically processed and released in response to cellular injury, leading to apoptosis.rs1800587 (−889 C/T)SNP[29]
IL1β2q14.1Encodes a member of the interleukin-1 cytokine family. This cytokine is produced by activated macrophages as a pro-protein, which is proteolytically processed to its active form by Caspase 1. It is an important mediator of the inflammatory response and is related to various cellular activities, including cell proliferation, differentiation, and apoptosis.rs16944
(−511 C/T)
SNP[30,31,32,33,34,35,36]
IL1RN2q14.1Encodes a member of the interleukin-1 cytokine family. This protein inhibits the activities of IL-1α and IL-1β and modulates various immune and inflammatory responses related to interleukin-1, particularly during the acute phase of infection and inflammation.(VNTR 86 bp)Variable number tandem repeat (VNTR)[30,31,37]
IL45q31.1Encodes a pleiotropic cytokine produced by activated T cells. IL-4, a type 2 cytokine, is considered important for tissue repair, counteracting the effects of pro-inflammatory type 1 cytokines. Additionally, it intervenes and regulates various responses in the human host, such as acute inflammation.rs2243250
(−590 C/T)
SNP[38,39]
IL67p15.3Encodes a cytokine that acts on inflammation and the maturation of B cells. This cytokine is primarily produced in sites of acute and chronic inflammation, where it is secreted into the serum and induces a transcriptional inflammatory response through the interleukin-6 alpha receptor.rs1800796
(−634 C/G)
SNP[40,41,42]
rs1800795 (−174 G/C)[43]
rs2069837 (A/G)[41]
rs1524107 (T/C)
(−176 G/C)[39]
IL8
(CXCL8)
4q13.3Encodes a protein that is a member of the CXC chemokine family; one of the main mediators of the inflammatory response. IL-8 acts as a chemotactic factor, directing neutrophils to the site of infection. It also participates, along with other cytokines, in the pro-inflammatory signaling cascade.rs4073
(−251 T/A)
SNP[17]
rs2227306 (+781 C/T)[44]
IL101q32.1Encodes a cytokine primarily produced by monocytes and to a lesser extent by lymphocytes. This cytokine has pleiotropic effects on immunoregulation and inflammation, and negatively regulates the expression of Th1 cytokines, MHC class II molecules, etc. It can block NF-kappa B activity and is related to the regulation of the JAK-STAT signaling pathway.rs1800896
(−1082 G/A)
SNP[45,46,47,48,49]
rs1800872
(−592 C/A)
[50]
IL3416q22.1Encodes a cytokine that promotes the differentiation and viability of monocytes and macrophages through the colony-stimulating factor 1 receptor (CSF1R).rs6499323 (G/A)SNP[51]
LTA6p21.33Encodes a protein member of the tumor necrosis factor family; a cytokine produced by lymphocytes. This protein also intervenes in various inflammatory, immune-stimulatory, and antiviral responses.rs1041981 (Thr26Asn; 804 C/A)SNP[52]
rs909253 (Ala252Gly; A/G)
MMP920q13.12Matrix metalloproteinases (MMPs) act on pro-inflammatory mediators, regulating various aspects of inflammation, and can function as a switch in acute and chronic inflammation. Therefore, MMP9 is considered a pro-inflammatory cytokine.rs17576
(Arg 279Gln, G/A)
SNP[17,53]
MPO17q22Myeloperoxidase (MPO) is a heme protein produced during myeloid differentiation, constituting an essential component of the azurophilic granules in neutrophils. This enzyme provides essential hypohalous acids for the microbicidal activity of neutrophils.rs2333227
(−463 G/A)
SNP[54,55]
NFKB14q24NF-κB is a transcription regulator activated by various intra- and extra-cellular stimuli, such as cytokines, free radicals, etc. When activated, it translocates to the nucleus and stimulates the expression of genes involved in various biological functions, such as cell growth and immune cell development.rs28362491
(−94 ATTG)
Indel[56]
NOS217q11.2Encodes the protein Nitric Oxide Synthase 2, which increases the synthesis of pro-inflammatory mediators, such as IL6 and IL8.rs2779248 (T/C)SNP[57]
rs1137933 (G/A)
OPN (SPP1)4q22.1Encodes a cytokine involved in increasing the production of interferon-gamma and interleukin-12 and reducing interleukin-10. This cytokine is essential in the pathway leading to type I immunity.rs11730582
(−443 C/T)
SNP[58]
PPARγ3p25.2Encodes a member of the nuclear receptor subfamily of the peroxisome proliferator-activated receptor (PPAR). PPAR-γ can inhibit the expression of pro-inflammatory genes, suppressing responses mediated by NF-kappa-B.rs1801282 (Pro12Ala; C/G)SNP[59]
pri-miR-125a19q13.41Encodes a miRNA considered a potential regulator of IL-6R. Li; Lei (2015) report that the expression level of the IL-6R protein was significantly decreased by the introduction of miR-125a mimetics.rs12976445 (C/T)SNP[60]
PRKCB116p12.2-p12.1Protein kinase Cs (PKCs) are a family of serine/threonine-specific protein kinases that plays an important role in B-cell activation by regulating B-cell receptor (BCR)-mediated NF-kappa-B activation.rs3760106
(−1504 C/T)
SNP[61]
rs2575390
(−546 C/G)
PTX33q25.32The expression of the protein produced by this gene is induced by inflammatory cytokines in response to inflammatory stimuli in various types of mesenchymal and epithelial cells. It promotes fibrocyte differentiation and is involved in the regulation of inflammation and complement activation.rs2305619
(281 A/G)
SNP[62,63]
RAGE (AGER)14q32.31Encodes a protein considered an intracellular signal transducer or pro-inflammatory peptide. It acts as a mediator of acute and chronic vascular inflammation, regulating the production/expression of TNF-alpha, oxidative stress, and endothelial dysfunction in type 2 diabetes.rs1800624
(−374 T/A)
SNP[64]
rs3134940
(2184 A/G)
[65]
RANTES (CCL5)17q12Produces a protein chemotactic for blood monocytes, memory helper T cells, and eosinophils. Additionally, it can activate various chemokine receptors, including CCR1, CCR3, CCR4, and CCR5.rs2280788
(−28 C/G)
SNP[20]
SASH16q24.3-q25.1Encodes a scaffold protein related to the TLR4 signaling pathway that can induce cytokine production and migration of endothelial cells in response to invading pathogens.rs6930576 (G/A)SNP[66]
SEPS1 (SELENOS)15q26.3Encodes a transmembrane protein located in the endoplasmic reticulum. It is involved in the process of degrading misfolded proteins and may play a role in inflammation control, acting as an anti-inflammatory and antioxidant protein.rs4975814 (G/T)SNP[67]
SLC12A316q13Encodes an electroneutral sodium and chloride ion cotransporter. It is a receptor for the pro-inflammatory cytokine IL18 and contributes to IL18-induced cytokine production, including IFNG, IL6, IL18, and CCL2.rs11643718 (Arg913Gln;
78 G/A)
SNP[68]
SUMO46q25.1Encodes small ubiquitin-related modifiers. The protein encoded by this gene particularly modifies IKBA, causing negative regulation of NF-kappa-B-dependent transcription of the IL12B gene.rs237025 (M55V; c.163 G/A)SNP[69]
TCRBC (TRB)7q34Encodes T cell receptors that recognize processed foreign antigens as small peptides bound to major histocompatibility complex molecules on the surface of antigen-presenting cells.(9.2;10.0 kb)Not identified[70]
TGFβ119q13.2Encodes a secreted ligand of the TGF-beta protein superfamily. This protein regulates cell proliferation, differentiation, and growth, and may modulate the expression and activation of other growth factors, such as interferon-gamma and tumor necrosis factor-alpha.rs1800470
(869 T/C)
SNP[71,72,73,74,75,76]
rs1800471
(915 G/C)
[74]
TLR49q33.1Encodes a protein member of the Toll-like receptor (TLR) family that plays a fundamental role in pathogen recognition and activation of innate immunity. They recognize molecular patterns associated with pathogens and are involved in the production of cytokines necessary for effective immunity.rs5030718
(14367 G/A)
SNP[77]
TNFα6p21.33Encodes a multifunctional pro-inflammatory cytokine belonging to the tumor necrosis factor superfamily. This cytokine is primarily secreted by macrophages and is involved in the regulation of various biological processes, such as cell proliferation, differentiation, apoptosis, lipid metabolism, and coagulation.rs1799964
(−1031 T/C)
SNP[34,78,79]
rs1800629
(−308 G/A)
[34,80,81]
rs361525
(−238 G/A)
[34]
TSC22 (TSC22D1)13q14.11Encodes a leucine zipper protein expressed in many tissues and involved in the signaling of TGF-1.(−396 A/G)SNP[73]
UMOD16p12.3Encodes a protein that can bind to immunoglobulin G, complement 1q, and tumor necrosis factor-alpha (TNF-α), signaling a role in innate immunity. It can also act as a receptor for the binding and endocytosis of cytokines (IL-1 and IL-2) and TNF.rs4293393 (T/C)SNP[82]
VEGF-A6p21.1Encodes a protein that acts as a pro-inflammatory cytokine, increasing the permeability of endothelial cells and inducing the expression of endothelial cell adhesion molecules through its ability to act as a chemotactic agent for monocytes.rs35569394
(−2549 D/I)
Indel[83]
Table 4. Twenty genes with the highest number of interactions according to CytoHubba.
Table 4. Twenty genes with the highest number of interactions according to CytoHubba.
RankingGeneFull Gene NameScore
1TNFTumor Necrosis Factor54.0
2IL1BInterleukin 1 Beta52.0
3IL6Interleukin 650.0
3IL10Interleukin 1050.0
6ICAM1Intercellular Adhesion Molecule 148.0
6CXCL8 (IL8)Chemokine (C-X-C motif) ligand 848.0
6MMP9Matrix Metallopeptidase 948.0
6TLR4Toll-like receptor 448.0
6VEGFAVascular Endothelial Growth Factor A48.0
11IL4Interleukin 446.0
12IL1AInterleukin 1 Alpha44.0
13CCL5Chemokine (C-C motif) ligand 542.0
14MPOMyeloperoxidase38.0
14ADIPOQAdiponectin, C1Q and Collagen Domain Containing38.0
14TGFB1Transforming Growth Factor Beta 138.0
17LTALymphotoxin Alpha36.0
18SPP1Secreted Phosphoprotein 134.0
18IL1RNInterleukin 1 Receptor Antagonist34.0
920NOS2Nitric Oxide Synthase 232.0
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Costa, C.C.P.d.; Assunção, L.d.P.; Santos, K.d.F.; Silva, L.d.; Santos, R.d.S.; Reis, A.A.d.S. In Silico Characterization of Inflammatory and Anti-Inflammatory Modulation in Diabetic Nephropathy: The Construction of a Genetic Panel. J. Mol. Pathol. 2024, 5, 335-359. https://doi.org/10.3390/jmp5030024

AMA Style

Costa CCPd, Assunção LdP, Santos KdF, Silva Ld, Santos RdS, Reis AAdS. In Silico Characterization of Inflammatory and Anti-Inflammatory Modulation in Diabetic Nephropathy: The Construction of a Genetic Panel. Journal of Molecular Pathology. 2024; 5(3):335-359. https://doi.org/10.3390/jmp5030024

Chicago/Turabian Style

Costa, Caroline Christine Pincela da, Leandro do Prado Assunção, Kamilla de Faria Santos, Laura da Silva, Rodrigo da Silva Santos, and Angela Adamski da Silva Reis. 2024. "In Silico Characterization of Inflammatory and Anti-Inflammatory Modulation in Diabetic Nephropathy: The Construction of a Genetic Panel" Journal of Molecular Pathology 5, no. 3: 335-359. https://doi.org/10.3390/jmp5030024

Article Metrics

Back to TopTop