Open Access

Prediction of seed gene function in progressive diabetic neuropathy by a network‑based inference method

  • Authors:
    • Shan‑Shan Li
    • Xin‑Bo Zhao
    • Jia‑Mei Tian
    • Hao‑Ren Wang
    • Tong‑Huan Wei
  • View Affiliations

  • Published online on: March 26, 2019     https://doi.org/10.3892/etm.2019.7441
  • Pages: 4176-4182
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Guilt by association (GBA) algorithm has been widely used to statistically predict gene functions, and network‑based approach increases the confidence and veracity of identifying molecular signatures for diseases. This work proposed a network‑based GBA method by integrating the GBA algorithm and network, to identify seed gene functions for progressive diabetic neuropathy (PDN). The inference of predicting seed gene functions comprised of three steps: i) Preparing gene lists and sets; ii) constructing a co‑expression matrix (CEM) on gene lists by Spearman correlation coefficient (SCC) method and iii) predicting gene functions by GBA algorithm. Ultimately, seed gene functions were selected according to the area under the receiver operating characteristics curve (AUC) index. A total of 79 differentially expressed genes (DEGs) and 40 background gene ontology (GO) terms were regarded as gene lists and sets for the subsequent analyses, respectively. The predicted results obtained from the network‑based GBA approach showed that 27.5% of all gene sets had a good classified performance with AUC >0.5. Most significantly, 3 gene sets with AUC >0.6 were denoted as seed gene functions for PDN, including binding, molecular function and regulation of the metabolic process. In summary, we predicted 3 seed gene functions for PDN compared with non‑progressors utilizing network‑based GBA algorithm. The findings provide insights to reveal pathological and molecular mechanism underlying PDN.

Introduction

Diabetic neuropathy (DN) is the most common and troublesome complication of diabetes, characterized by the gradual loss of peripheral axons, resulting in diminished sensation, pain, and eventual complete loss of sensation (1). Pathologically, it is the culprit of a series of interrelated metabolic abnormalities with insulin deficiency and hyperglycemia (2). Importantly, DN affects up to 60–70% of diabetics, leading to the highest morbidity and mortality rates and to a huge economic burden of diabetes care (3,4). However, in addition to controlling blood glucose levels, no effective treatment options have been found to prevent, slow or reverse the progression of DN and are not always achievable even in alert patients (5). Besides, a good understanding of pathological and molecular mechanism underlying DN might give help to explore effective therapy of this complicated disease.

The difference of gene expression levels could reflect the propensity of many diseases, and thus identifying gene functions has been an effective way to reveal the pathological mechanism of a disease at molecular level (6). ELAVL3, as a member of the Elavl family, is known as a neuronal Elavl because it is expressed in peripheral and central neurons throughout development (7). Ogawa et al hold the view that ELAVL3 is essential for maintaining Purkinje neuron axons (8). It has been reported that ELAVL3 regulates neuronal polarity via alternative splicing of embryo-specific exon in AnkyrinG (9). There is some evidence to suggest that Molecule Interacting with CasL-Like1 (MICALL1), involved in pre-cytokinetic events, acts as a membrane hub on tubular recycling endosomes (10). According to recent reports, tubular recirculating endosome biogenesis was regulated through p53-MICALL1 pathway (11). HEY2, as a member of hairy-related basic helix-loop-helix transcription factor subfamily, is involved in boundary formation and cell fate determination (12). Recent research has indicated that miR-98 activated the Notch signaling pathway by binding to HEY2 in Alzheimer's disease mice to improve mitochondrial dysfunction and oxidative stress (13). Our research results agree with previous studies, suggesting that ELAVL3, MICALL1, HEY2 genes have effects on neurological diseases. It has been demonstrated that by using variants based on the guilty susceptibility (GBA) algorithm, gene function prediction can be performed with very high statistical confidence assuming that the associations in the genetic data is necessary to establish culpability (14). Although various of techniques have been proposed for purpose of extending the GBA to indirect connections, only slight effectiveness has been discovered (1518).

In the present study, a new method was proposed to predict seed gene functions for progressive DN (PDN) patients, by integrating the GBA algorithm and network-based method. To achieve this goal, gene expression data and gene ontology (GO) annotations were collected from the public databases. Differentially expressed genes (DEGs) were identified as gene lists and background GO terms were extracted as gene sets. The co-expression matrix (CEM) was constructed on gene lists by Spearman correlation coefficient (SCC) method.

Materials and methods

Preparing gene expression data

Gene expression data [GSE24290 (19)] for human DN were recruited from the public-free Gene Expression Omnibus (GEO). In brief, based on the density of myelinated fibers, GSE24290 divided the DN patient samples into two groups, progressors and non-progressors. In short, the patients in the progressive group lost ≥500 fibres/mm2, and the patients in the non-progressive group lost ≤100 fibres/mm2 over 52 weeks (20). By mapping these preprocessed probes to gene structures, a total of 10,570 genes were identified for PDN for subsequent analysis.

Collecting gene sets

All human GO annotations were prepared from the Gene Ontology Consortium (21,22). For purpose of making these retained GO terms more correlated to progressors, we took the intersections between DEGs and GO terms. If the number of DEGs for a GO term was smaller than 20, it was removed. Only GO terms including equal or more than 20 DEGs were reserved.

Identifying DEGs

DEGs between progressors and non-progressors were detected (23). The lmFit function implemented in Limma was utilized to perform linear fitting, empirical Bayes statistics and false discovery rate (FDR) calibration of the P-values on the data (24,25). The thresholds for DEGs were set as P<0.05 and |log2 fold-change| >2.

Constructing CEM

To further investigate the correlations or interactions among DEGs, a CEM for them was constructed based on the SCC algorithm (26). If the SCC for a pair of genes was positive, it would indicate a positive linear correlation between the two genes. Similarly, a negative SCC refers to a negative relationship of the gene pair. In addition, the absolute SCC value of an interaction was denoted as its weight value. Furthermore, the higher the weight value across two genes, the stronger the interaction was, especially for 1. Otherwise, the 0 meant that there was no interaction between two genes. As a result, a CEN was constructed according to weight.

Network-based GBA algorithm

The GBA algorithm was combined with network to predict significant gene functions for progressors. Specifically, for a DEG in the CEM, we chose its adjacent genes to enrich to a GO term. Based on these GO functional annotations, a multi-functionality (MF) score was assigned to each gene i in the CEM (14)

S(i)=∑k∨i∈GOk1Numink*Numoutk

of which Numink represented the number of genes within GO group k, whose weighting had the effect of giving contribution to a GO group; and Numoutk was the number of genes outside GO group k in the CEM, whose weighting provided a corresponding weighting to genes not within the GO group. Note that we computed the AUC for evaluating classification performances between progressors and non-progressors. Here, for assessing the predictive power of machine learners in the support vector machine (SVM) model, AUC is an assessment of the accuracy of clinical classification performance (27). Most importantly, an AUC of 0.5 represents classification at chance levels, while an AUC of 1.0 represents a perfect classification. Thus we defined GO terms of AUC >0.6 as seed gene functions for PDN patients in the present report.

Results

Gene lists

In this report, based on 10,570 genes in GSE24290, we identified 79 DEGs between progressors and non-progressors by Limma package when setting the thresholds as P<0.05 and |log2 fold-change| >2. All DEGs were ranked in ascending order of their P-values (Table I). We found that the most significant DEGs were ELAVL3 (P=1.87E-02), MICALL1 (P=2.51E-02), HEY2 (P=3.42E-02), PCDHB1 (P=3.69E-02) and OR2S2 (P=4.81E-02). Importantly, the 79 DEGs were regarded as gene lists for further exploitation.

Table I.

Gene list for progressive diabetic neuropathy (PDN).

Table I.

Gene list for progressive diabetic neuropathy (PDN).

RankDEGP-valueRankDEGP-value
1ELAVL30.01873014941FER1L40.237218317
2MICALL10.02506416842TAS2R130.254492341
3HEY20.03418301143SYN10.254519864
4PCDHB10.03693842144KCNMB40.257387331
5OR2S20.04811265245SLC35E10.258472445
6CNTD20.05597784246PRX0.267184163
7DNAJB120.05921390347GIPC20.276805505
8GPR210.06109171248RBM12B0.298659702
9PFN10.06835143649HACD10.302339336
10ZMIZ20.07394446250NAALAD20.309726354
11HSPA60.08046779451KLHL210.310864132
12TRIM360.08292292552TMX20.316649651
13MTHFD10.0831372653HEBP20.319262815
14RPA40.09127762854RFC20.319345713
15NIPAL20.10102455855CCDC1340.327623954
16HDDC20.10618130456MEPE0.343481838
17LIME10.10710138357TRAPPC90.374991751
18MAPKAPK5-AS10.10886769158TXNRD10.384530053
19CCDC1860.11657363659CTDNEP10.386873156
20FCF10.11756987560SH2D4A0.39096557
21PSMD50.11864195261ZNF512B0.390996512
22YTHDF30.13085380762TAGLN20.400497742
23CDHR50.13595299163C10orf760.401347031
24C6orf1200.141611564PIK3IP10.409666004
25VWA70.14417817865C5orf300.420802441
26ASF1A0.14720845766PKNOX10.449555958
27NELL20.16223325267DNAJB20.465398033
28SRM0.16754558668NCR20.474076273
29SH3KBP10.17645949369GALNT80.475841943
30USE10.17899328770IKZF30.480703371
31INSIG10.17939717171SENP50.492341515
32SSH30.1832818772POMGNT20.510089318
33FAM66D0.18533387473CALHM20.515258516
34PGAP30.19087942474ATP8B40.5305693
35PER20.19265545575GALNT100.543825962
36CCS0.19310632176WIZ0.550465591
37E2F80.20007461477FER0.594006462
38HTR1F0.22184032478PITPNA0.597706177
39ZNF1420.23230106979MYO3A0.599378182
40PPP1R3B0.236879137
Gene sets

In the Gene Ontology Consortium database, there are 19,003 gene sets involved in 18,402 genes. To make these sets with a stable performance and more correlated to progressors, we chose GO terms from 20 to 1,000 in size and then took intersections between the reserved terms with the gene lists. Only gene sets containing intersected DEGs >20 were left in the subsequent analysis. Hereinafter, we defined the amount of intersected DEGs as the count value of this term. As a result, a total of 40 GO terms were determined (Table II), termed with background GO terms. There were three terms with Count >60, cellular component (GO:0005575, Count = 71), biological process (GO:0008150, Count = 67), and molecular function (GO:0003674, Count = 66). Furthermore, we calculated the DEGs by expressing the spectral data. According to the GO enrichment analysis, the enrichment of 79 DEGs in background GO terms was obtained (28). DEG annotated to a GO term, and if the gene is present in the GO term, the value is 1 (red), otherwise 0 (yellow). Finally, the heatmap was obtained from the heatmap package in the R language to analyze the above enrichment situation (Fig. 1). The result of this heatmap was in accordance with Table II. Ultimately, the background GO terms were the gene sets used.

Table II.

Gene sets for progressive diabetic neuropathy (PDN).

Table II.

Gene sets for progressive diabetic neuropathy (PDN).

GO IDGO termCountsGO IDGO termCounts
GO:0005575Cellular component71GO:0008152Metabolic process36
GO:0008150Biological process67GO:0071704Organic substance metabolic process34
GO:0003674Molecular function66GO:0016020Membrane33
GO:0005623Cell58GO:0044237Cellular metabolic process33
GO:0044464Cell part58GO:0044238Primary metabolic process33
GO:0009987Cellular process57GO:0043170Macromolecule metabolic process28
GO:0005488Binding55GO:0044425Membrane part27
GO:0044699Single-organism process52GO:0044444Cytoplasmic part27
GO:0043226Organelle49GO:0044260Cellular macromolecule metabolic process26
GO:0044763Single-organism cellular process49GO:0005634Nucleus25
GO:0005622Intracellular47GO:0016021Integral component of membrane25
GO:0044424Intracellular part47GO:0031224Intrinsic component of membrane25
GO:0043227Membrane-bounded organelle46GO:0043167Ion binding24
GO:0065007Biological regulation43GO:0050896Response to stimulus24
GO:0043229Intracellular organelle42GO:0044422Organelle part23
GO:0005515Protein binding41GO:0097159Organic cyclic compound binding23
GO:0043231Intracellular membrane-bounded organelle40GO:1901363Heterocyclic compound binding23
GO:0050789Regulation of biological process39GO:0044446Intracellular organelle part22
GO:0005737Cytoplasm38GO:0019222Regulation of metabolic process21
GO:0050794Regulation of cellular process37GO:0032502Developmental process21
CEM

With an attempt to investigate biological correlations among DEGs, a CEM with 79 nodes and 3,081 interactions were constructed based on the SCC, of which each interaction possessed a weight value to reveal the interacted strength between two genes. The weight distribution for interactions in this CEM showed the characteristic of good adjacent matrix that weights on its diagonal nearly equaled to 1, which suggested that the CEM had a good network scale property. In particular, an edge between C6orf120 and PGAP3 had the highest weight of 0.994. Moreover, to further evaluate the activities of genes in interactions of high weights, topological degree centrality analysis was conducted on all nodes in the CEM. The results showed that C6orf120 connected with 56 adjacent genes and thus had the highest degree. Subsequently, an assortativity coefficient was calculated to assess degree assortative mixing pattern extent. Consequently, the assortativity coefficient for the CEM was 0.829, indicating that the network had perfect assortative mixing patterns.

A sub-network was extracted from the CEM by selecting those interactions with weight >0.8 and visualized it by Cytoscape software (Fig. 2). There were 48 nodes and 330 edges in the sub-network. Among these nodes, PGAP3, C6orf120 and RBM12B had higher degree than the others, which suggested their key roles in the PCN patients and inferred that gene functions participated by them might act as critical processes in progressors.

Seed gene functions

The AUC index was applied to evaluate the classified performance on MF scores between progressors and non-progressors by the 3-fold cross-validation. Generally, if an AUC for a gene or process was >0.5, it could be used to classify the case group from controls. In this report, the AUC for the 40 gene sets are shown in Fig. 3. There were 11 GO terms with AUC >0.5 in total. Thus, 11 of 40 (27.5%) gene sets had a good classification performance between progressors and non-progressors of DN. Among them, 3 with AUC >0.6 were considered to be seed gene functions in progressors of DN. The seed gene functions were Binding (GO:0005488, AUC=0.668), Molecular function (GO:0003674, AUC=0.654), and Regulation of metabolic process (GO:0019222, AUC=0.636).

Discussion

In the present study, we predicted seed gene functions in PDN patients by using a network-based GBA method, since the network-based approach systematically investigate the molecular complexity of a particular disease (29) and identify potential signatures through bio-molecular networks rather than individual genes (30,31). An integration of co-expression network and the GBA algorithm provide a new manner to predict significant gene functions and reveal molecular mechanism underlying PDN.

On the basis of SCC method, a CEM for progressors was constructed on DEGs, and a sub-network of weight >0.8 was extracted from the CEM. Interestingly, we found that PGAP3, C6orf120 and RBM12B had high degree both in CEM and its sub-network, which indicated their importance in progressors. Taking RBM12B as an example, RBM12B (RNA binding motif protein 12B) is a protein coding gene that relates to functions of RNA binding, nucleic acid binding and nucleotide binding (32). It has been demonstrated that Rbm12b and Rbm3 are mainly down-regulated and highly responsive to systemic hypoxia in mouse developing brain and placenta (33). This is the first time the key role of RBM12B in human PDN patients was uncovered. In addition, in our study, this gene was enriched in two seed gene functions, Binding and Molecular function. The possible inference was that the dys-regulation of RBM12B might disturb the normal functions of binding and lead to brain injury, even lesions of the nervous system.

Particularly, a total of 40 background GO terms were identified as gene sets for the current study. Subsequently, an MF score was assigned to each gene in the specific gene set, and then an AUC for each GO term was produced to assess the prediction performance between progressors and non-progressors. In consequence, 27.5% of all gene sets had a good classified performance with AUC >0.5. Most significantly, 3 gene sets with AUC >0.6 were denoted as seed gene functions for PDN, including Binding, Molecular function and Regulation of metabolic process. It is a common phenomenon that a given gene is present in one or more molecular functions (such as RBM12B described above), and two or more genes exhibit the same function.

Cameron et al reviewed that poor metabolic control was one of microvascular complications correlated to DN (34). Thus, it was important to improve the metabolic conditions that led to the pathology underlying peripheral DN, and the metabolic correction modified symptom control and clinical results (35). Moreover, metabolic dysfunction in experimental DN due to energy homeostasis and/or oxidative stress is limited to the sciatic nerve (36). Above all, we might conclude various metabolic processes play crucial roles in DP, and thus the regulation of metabolic process has become informative in PDN patients.

In summary, we have predicted 3 seed gene functions for progressors of DP compared with non-progressors utilizing network-based GBA algorithm. The findings provide insights to reveal pathological and molecular mechanism underlying PDN. However, the expression data used in this work was recruited from the open access database, and the 3 seed gene functions still need to be validated.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

SSL and XBZ conceived the study and drafted the manuscript. XBZ, JMT and HRW acquired the data. SSL and THW analyzed the data and revised the manuscript. All the authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Edwards JL, Vincent AM, Cheng HT and Feldman EL: Diabetic neuropathy: Mechanisms to management. Pharmacol Ther. 120:1–34. 2008. View Article : Google Scholar : PubMed/NCBI

2 

Sima AAF and Zhang W: Mechanisms of diabetic neuropathy: Axon dysfunction. Handb Clin Neurol. 126:429–442. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Vinik AI, Nevoret ML, Casellini C and Parson H: Diabetic neuropathy. Endocrinol Metab Clin North Am. 42:747–787. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Oyenihi AB, Ayeleso AO, Mukwevho E and Masola B: Antioxidant strategies in the management of diabetic neuropathy. BioMed Res Int. 2015:5150422015. View Article : Google Scholar : PubMed/NCBI

5 

Charnogursky G, Lee H and Lopez N: Diabetic neuropathy. Handb Clin Neurol. 120:773–785. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Zhao J, Yang TH, Huang Y and Holme P: Ranking candidate disease genes from gene expression and protein interaction: A Katz-centrality based approach. PLoS One. 6:e243062011. View Article : Google Scholar : PubMed/NCBI

7 

Good PJ: The role of elav-like genes, a conserved family encoding RNA-binding proteins, in growth and development. Semin Cell Dev Biol. 8:577–584. 1997. View Article : Google Scholar : PubMed/NCBI

8 

Ogawa Y, Kakumoto K, Yoshida T, Kuwako KI, Miyazaki T, Yamaguchi J, Konno A, Hata J, Uchiyama Y, Hirai H, et al: Elavl3 is essential for the maintenance of Purkinje neuron axons. Sci Rep. 8:27222018. View Article : Google Scholar : PubMed/NCBI

9 

Ogawa Y, Yamaguchi J, Yano M, Uchiyama Y and Okano HJ: Elavl3 regulates neuronal polarity through the alternative splicing of an embryo-specific exon in AnkyrinG. Neurosci Res. 135:13–20. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Reinecke JB, Katafiasz D, Naslavsky N and Caplan S: Novel functions for the endocytic regulatory proteins MICAL-L1 and EHD1 in mitosis. Traffic. 16:48–67. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Takahashi Y, Tanikawa C, Miyamoto T, Hirata M, Wang G, Ueda K, Komatsu T and Matsuda K: Regulation of tubular recycling endosome biogenesis by the p53-MICALL1 pathway. Int J Oncol. 51:724–736. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Miao L, Li J, Li J, Tian X, Lu Y, Hu S, Shieh D, Kanai R, Zhou BY, Zhou B, et al: Notch signaling regulates Hey2 expression in a spatiotemporal dependent manner during cardiac morphogenesis and trabecular specification. Sci Rep. 8:26782018. View Article : Google Scholar : PubMed/NCBI

13 

Chen FZ, Zhao Y and Chen HZ: MicroRNA-98 reduces amyloid β-protein production and improves oxidative stress and mitochondrial dysfunction through the Notch signaling pathway via HEY2 in Alzheimer's disease mice. Int J Mol Med. 43:91–102. 2019.PubMed/NCBI

14 

Gillis J and Pavlidis P: The impact of multifunctional genes on ‘guilt by association’ analysis. PLoS One. 6:e172582011. View Article : Google Scholar : PubMed/NCBI

15 

Chua HN, Sung WK and Wong L: Exploiting indirect neighbours and topological weight to predict protein function from protein-protein interactions. Bioinformatics. 22:1623–1630. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Yip AM and Horvath S: Gene network interconnectedness and the generalized topological overlap measure. BMC Bioinformatics. 8:222007. View Article : Google Scholar : PubMed/NCBI

17 

Weston AD and Hood L: Systems biology, proteomics, and the future of health care: Toward predictive, preventative, and personalized medicine. J Proteome Res. 3:179–196. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Peña-Castillo L, Tasan M, Myers CL, Lee H, Joshi T, Zhang C, Guan Y, Leone M, Pagnani A, Kim WK, et al: A critical assessment of Mus musculus gene function prediction using integrated genomic evidence. Genome Biol. 9 (Suppl 1):S22008. View Article : Google Scholar

19 

Hur J, Sullivan KA, Pande M, Hong Y, Sima AA, Jagadish HV, Kretzler M and Feldman EL: The identification of gene expression profiles associated with progression of human diabetic neuropathy. Brain. 134:3222–3235. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Wiggin TD, Sullivan KA, Pop-Busui R, Amato A, Sima AAF and Feldman EL: Elevated triglycerides correlate with progression of diabetic neuropathy. Diabetes. 58:1634–1640. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Consortium GO; Gene Ontology Consortium, : Gene Ontology Consortium: Going forward. Nucleic Acids Res. 43(D1): D1049–D1056. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Gillis J and Pavlidis P: The role of indirect connections in gene networks in predicting function. Bioinformatics. 27:1860–1866. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

24 

Datta S, Satten GA, Benos DJ, Xia J, Heslin MJ and Datta S: An empirical bayes adjustment to increase the sensitivity of detecting differentially expressed genes in microarray experiments. Bioinformatics. 20:235–242. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Reiner A, Yekutieli D and Benjamini Y: Identifying differentially expressed genes using false discovery rate controlling procedures. Bioinformatics. 19:368–375. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Szmidt E and Kacprzyk J: The Spearman rank correlation coefficient between intuitionistic fuzzy sets. 2010 5th IEEE International Conference Intelligent Systems. IEEE; London: pp. 276–280. 2010, View Article : Google Scholar

27 

Huang J and Ling CX: Using AUC and accuracy in evaluating learning algorithms. Knowledge and Data Engineering. IEEE Trans. 17:299–310. 2005.

28 

Zhu Q, Sun Y, Zhou Q, He Q and Qian H: Identification of key genes and pathways by bioinformatics analysis with TCGA RNA sequencing data in hepatocellular carcinoma. Mol Clin Oncol. 9:597–606. 2018.PubMed/NCBI

29 

Barabási AL, Gulbahce N and Loscalzo J: Network medicine: A network-based approach to human disease. Nat Rev Genet. 12:56–68. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Liu ZP, Wang Y, Zhang XS and Chen L: Network-based analysis of complex diseases. IET Syst Biol. 6:22–33. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Chen L, Wang RS and Zhang XS: Reconstruction of gene regulatory networks. In: Biomolecular Networks: Methods and Applications in Systems Biology. John Wiley & Sons Inc.; Hoboken, NJ: pp. 47–87. 2009

32 

Zhang G, Bowling H, Hom N, Kirshenbaum K, Klann E, Chao MV and Neubert TA: In-depth quantitative proteomic analysis of de novo protein synthesis induced by brain-derived neurotrophic factor. J Proteome Res. 13:5707–5714. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Trollmann R, Rehrauer H, Schneider C, Krischke G, Huemmler N, Keller S, Rascher W and Gassmann M: Late-gestational systemic hypoxia leads to a similar early gene response in mouse placenta and developing brain. Am J Physiol Regul Integr Comp Physiol. 299:R1489–R1499. 2010. View Article : Google Scholar : PubMed/NCBI

34 

Cameron NE, Eaton SEM, Cotter MA and Tesfaye S: Vascular factors and metabolic interactions in the pathogenesis of diabetic neuropathy. Diabetologia. 44:1973–1988. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Miranda-Massari JR, Gonzalez MJ, Jimenez FJ, Allende-Vigo MZ and Duconge J: Metabolic correction in the management of diabetic peripheral neuropathy: Improving clinical results beyond symptom control. Curr Clin Pharmacol. 6:260–273. 2011. View Article : Google Scholar : PubMed/NCBI

36 

Freeman OJ, Unwin RD, Dowsey AW, Begley P, Ali S, Hollywood KA, Rustogi N, Petersen RS, Dunn WB, Cooper GJ, et al: Metabolic dysfunction is restricted to the sciatic nerve in experimental diabetic neuropathy. Diabetes. 65:228–238. 2016.PubMed/NCBI

Related Articles

Journal Cover

May-2019
Volume 17 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li SS, Zhao XB, Tian JM, Wang HR and Wei TH: Prediction of seed gene function in progressive diabetic neuropathy by a network‑based inference method. Exp Ther Med 17: 4176-4182, 2019
APA
Li, S., Zhao, X., Tian, J., Wang, H., & Wei, T. (2019). Prediction of seed gene function in progressive diabetic neuropathy by a network‑based inference method. Experimental and Therapeutic Medicine, 17, 4176-4182. https://doi.org/10.3892/etm.2019.7441
MLA
Li, S., Zhao, X., Tian, J., Wang, H., Wei, T."Prediction of seed gene function in progressive diabetic neuropathy by a network‑based inference method". Experimental and Therapeutic Medicine 17.5 (2019): 4176-4182.
Chicago
Li, S., Zhao, X., Tian, J., Wang, H., Wei, T."Prediction of seed gene function in progressive diabetic neuropathy by a network‑based inference method". Experimental and Therapeutic Medicine 17, no. 5 (2019): 4176-4182. https://doi.org/10.3892/etm.2019.7441