Open Access

Gene microarray analyses for potential biomarkers of single and recurrent venous thromboembolism

  • Authors:
    • Wugang Zhou
    • Ke Zhang
    • Dongrui Chen
    • Pingjin Gao
    • Qiao Wang
  • View Affiliations

  • Published online on: September 22, 2015     https://doi.org/10.3892/mmr.2015.4349
  • Pages: 7358-7366
  • Copyright: © Zhou et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Venous thromboembolism is a major cause of morbidity and mortality with a high recurrence rate. The present study aimed to explore the molecular mechanisms and potential biomarkers of single venous thromboembolism (SVTE) and recurrent venous thromboembolism (RVTE). The microarray dataset GSE19151 was downloaded from Gene Expression Omnibus, which contained data from whole blood samples from 63 healthy controls, 32 SVTE and 38 RVTE patients. Differentially expressed genes (DEGs) in the SVTE and RVTE groups compared with those in the controls were identified using the t‑test, followed by clustering analysis of DEGs and samples. Functional and pathway enrichment analyses were performed for DEGs in patients with RVTE and SVTE, as well as specific DEGs in patients with RVTE. The identified 42 DEGs in RVTE were mainly enriched in biological processes of cellular protein metabolism, gene expression and translational elongation as well as in pathways associated with ribosomes, Parkinson's disease and oxidative phosphorylation. In SVTE, 20 DEGs were identified, which were mainly involved in biological processes of biopolymer biosynthesis, translational elongation and cellular protein metabolism as well as pathways associated with ribosomes and cardiac muscle contraction. In RVTE, 22 specific DEGs were mainly involved in translational elongation, negative regulation of the force of heart contraction by chemical signals, cell proliferation, ribosomal pathways and protein export. The identified DEGs of SVTE, including COX7C and UQCRQ, may be potential biomarkers for SVTE, and the specific DEGs of RVTE, including ADRBK1, NDUFA5 and ATP5O, may be potential biomarkers for RVTE.

Introduction

Venous thromboembolism (VTE), comprising deep venous thrombosis and pulmonary embolism, is a frequent disease with high morbidity and mortality, which affects 1–2 per 1,000 individuals (13). Furthermore, VTE is associated with a significant rate of recurrence in as many as 30% of VTE patients after termination of the standard course of anti-coagulant therapy (46). Therefore, it is desired to explore the molecular mechanisms and potential biomarkers that enable clinicians to identify patients at a risk of single VTE (SVTE) or recurrent VTE (RVTE) for prompt clinical diagnosis and early prevention (7).

Kyrle et al (8) reported that patients with a high level of factor VIII have an increased risk of RVTE. A study by Comp and Esmon (9) suggested that the levels of protein S may be used in the evaluation of RVTE. Various established and novel biomarkers, including D-dimer, E-selectin, P-selectin, thrombin, inflammatory markers and C-reactive protein, have been investigated for their predictive value in SVTE and RVTE (1013). However, only a small number of biomarkers, such as D-dimer, associated with a first or recurrent event of VTE were highlighted by these studies, while novel and promising biomarkers, including P-selectin and inflammatory cytokines, are still controversial (1). A study by Lewis et al (14) performed a pathway enrichment analysis of differentially expressed genes (DEGs) in samples from patients with SVTE and samples from patients with RVTE and found that insulin-like growth factor receptor 1 and Akt pathways may be useful for distinguishing patients with SVTE from those with RVTE.

The present study identified DEGs in RVTE and SVTE, as well as specific DEGs in RVTE. Functional and pathway enrichment analyses for these DEGs were performed to explore the molecular mechanisms and potential biomarkers of SVTE and RVTE in order to facilitate the diagnosis and clinical therapy management of VTE.

Materials and methods

Affymetrix microarray data

The gene expression profile dataset GSE19151 was obtained from Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/), which was deposited by Lewis et al (14). Microarray data from 133 whole blood specimens were available, including 63 samples from healthy controls, 32 samples from patients with SVTE (sampled at <three years since their most recent VTE) and 38 samples from subjects with recurrent venous thromboembolism (RVTE) on warfarin. The platform was GPL571 (HG-U133A_2) Affymetrix Human Genome U133A 2.0 Array (Affymetrix, Inc., Santa Clara, CA, USA).

DEG analysis and gene clustering analysis

For the GSE19151 dataset, the Bioconductor software package in R (http://www.bioconductor.org/; version 3.1) was implemented to analyze the 133 blood gene chips (15). Background correction and quartile data normalization were performed using the robust multiarray average algorithm with defaulted parameters in the Affy package (http://www.bioconductor.org/packages/release/bioc/html/affy.html; version 1.46.1) (16). The t-test was used to identify DEGs using the Simpleaffy package (http://www.bioconductor.org/packages/release/bioc/html/simpleaffy.html; version 2.44.0) (17). The DEGs were selected with the cutoff criteria of P<0.05 and |log(fold change)|>1. Hierarchical clustering analysis of the DEGs was performed using the Hclust command in R and the default complete linkage method (18).

Gene ontology (GO) functional and pathway enrichment analyses

The Integrated GEne and PROtein annotation Server (IGEPROS; http://www.biosino.org/iGepros/index.jsp) (19) bioinformatics resources consist of an integrated biological knowledge base and analytic tools aimed at systematically extracting biological information from large gene or protein lists. IGEPROS was used to perform the GO (http://www.geneontology.org/) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.genome.jp/kegg/pathway.html) pathway enrichment analyses for the identified DEGs with the threshold of P<0.05. The pathview package in R was utilized to depict the KEGG pathway (20).

Results

DEG selection and hierarchical clustering analysis

A total of 42 DEGs were identified between RVTE and normal whole-blood specimens (RVTE vs. control), including 35 up- and 7 downregulated genes. Subsequently, 20 DEGs between SVTE and normal whole-blood specimens (SVTE vs. control) were identified, including 17 up- and 3 downregulated genes. A total of 22 non-overlapping genes were selected as specific DEGs of RVTE, including 18 up- and 4 downregulated genes (Table I). Hierarchical clustering analysis was performed for the 42 DEGs from the 133 whole blood specimens of patients with SVTE, patients with RVTE and healthy controls. The result of this clustering analysis suggested that these DEGs may have important roles in VTE (Fig. 1).

Table I

Specific differentially expressed genes of recurrent VTE.

Table I

Specific differentially expressed genes of recurrent VTE.

Probe IDMean of recurrent VTE (R)Mean of healthy (N)Log2 (R/N)P-valueGene symbolENTREZGENE_ID
201304_at6.224.641.58 2.55×10−5NDUFA54698
200679_x_at9.868.671.19 4.85×10−10HMGB13146
201317_s_at9.498.161.33 2.18×10−7PSMA25683
200977_s_at9.418.161.24 1.39×10−5TAX1BP18887
201152_s_at11.8610.691.17 1.17×10−6MBNL14154
201163_s_at8.797.541.24 1.19×10−12IGFBP73490
201593_s_at9.388.261.12 3.27×10−10ZC3H1555854
201401_s_at8.579.69−1.13 1.39×10−11ADRBK1156
200834_s_at12.5411.541.01 1.97×10−11RPS216227
201367_s_at8.9110.14−1.24 2.12×10−7ZFP36L2678
201392_s_at9.0910.22−1.13 5.39×10−10IGF2R3482
201200_at10.989.821.16 1.72×10−11CREG18804
200012_x_at13.0811.661.42 1.40×10−6RPL216144
200061_s_at11.349.941.40 4.02×10−9RPS246229
200741_s_at13.5612.341.22 1.28×10−11RPS276232
200880_at8.927.731.19 1.17×10−7DNAJA13301
201012_at10.849.291.55 2.33×10−9ANXA1301
201273_s_at10.769.691.070.000332SRP96726
201332_s_at8.229.35−1.13 1.44×10−13STAT66778
201406_at10.359.031.32 6.59×10−5RPL36A6173
201023_at9.938.761.170.000185TAF76879
200818_at10.179.161.02 5.51×10−5ATP5O539

[i] VTE, venous thromboembolism; R, recurrent; N, normal.

GO enrichment analysis of DEGs

GO enrichment analysis was performed for 42 DEGs in RVTE, 20 DEGs in SVTE and 22 specific DEGs of RVTE. In RVTE, most enriched GO terms of DEGs in biological processes were associated with biopolymer biosynthesis, including cellular protein metabolism (P=1.12×10−8), gene expression (P=1.36×10−6), translational elongation (P=9.65×10−27) and cellular macromolecular biosynthetic processes (P=8.78×10−6). In the cellular component category, enriched GO terms were mainly associated with the ribosomal sub-unit (P=2.03×10−18), cytosol (P=2.43×10−12) and macromolecular complexes (P=8.61×10−11). In the molecular function category, GO terms enriched for DEGs in RVTE included structural constituents of ribosomes (P=9.13×10−24), insulin-like growth factor binding (P=0.002) and beta-adrenergic receptor kinase activity (P=0.005) (Tables IIIV).

Table II

GO functional enrichment analysis of DEGs in patients with RVTE and SVTE, and top 10 specific DEGs of RVTE associated with biological processes.

Table II

GO functional enrichment analysis of DEGs in patients with RVTE and SVTE, and top 10 specific DEGs of RVTE associated with biological processes.

Category/GOBPIDP-valueCount (n)Size (n)Term
RVTE
 GO:0006414 9.65×10−2717104Translational elongation
 GO:0044267 1.12×10−8222361Cellular protein metabolic process
 GO:0010467 1.36×10−6253592Gene expression
 GO:0043284 2.88×10−6233183Biopolymer biosynthetic process
 GO:0034645 8.78×10−6233386Cellular macromolecule biosynthetic process
 GO:0044237 1.49×10−5347160Cellular metabolic process
 GO:00090580.000114244223Biosynthetic process
 GO:00422740.000360210Ribosomal small subunit biogenesis
 GO:00442380.001536317368Primary metabolic process
 GO:00063640.002059388rRNA processing
SVTE
 GO:0006414 7.47×10−2212106Translational elongation
 GO:0044267 1.24×10−7142499Cellular protein metabolic process
 GO:0043284 2.35×10−5133183Biopolymer biosynthetic process
 GO:0034645 4.76×10−5133386Cellular macromolecule biosynthetic process
 GO:0010467 9.42×10−5113052Gene expression
 GO:00442370.000402177160Cellular metabolic process
 GO:00090580.000549134223Biosynthetic process
 GO:00066100.00400313Ribosomal protein import into nucleus
 GO:00063640.006046288rRNA processing
 GO:00422730.01064118Ribosomal large subunit biogenesis
Non-overlap
 GO:0006414 3.52×10−75104Translational elongation
 GO:00002960.00154711Spermine transport
 GO:00031080.00154711Negative regulation of the force of heart contraction by chemical signal
 GO:00082830.0021576917Cell proliferation
 GO:00442670.00294192361Cellular protein metabolic process
 GO:00069160.0029983190Anti-apoptosis
 GO:00485230.00303371334Negative regulation of cellular process
 GO:00459880.00309212Negative regulation of striated muscle contraction
 GO:00459000.00309212Negative regulation of translational elongation
 GO:00482950.00309212Positive regulation of isotype switching to IgE isotypes

[i] GO, gene ontology; DEG, differentially expressed genes; RVTE, recurrent venous thromboembolism; SVTE, single venous thromboembolism; non-overlap, non-overlapping DEGs as specific DEGs of RVTE; GOBPID, gene ontology biological process identification number; IgE, immunoglobulin E; Count, DEG numbers enriched in the specific GO term; Size, total gene numbers in the GO database.

Table IV

GO functional enrichment analysis of DEGs in patients with RVTE and SVTE, and top 10 specific DEGs of RVTE associated with molecular function.

Table IV

GO functional enrichment analysis of DEGs in patients with RVTE and SVTE, and top 10 specific DEGs of RVTE associated with molecular function.

Category/GOMFIDP-valueCount (n)Size (n)Term
RVTE
 GO:0003735 9.13×10−2417158Structural constituent of ribosome
 GO:0003723 2.87×10−610646RNA binding
 GO:00055200.002031225Insulin-like growth factor binding
 GO:00152660.00268611Protein channel activity
 GO:00150770.0029423107Monovalent inorganic cation transmembrane transporter activity
 GO:00476960.00536512Beta-adrenergic receptor kinase activity
 GO:00050100.00803713Insulin-like growth factor receptor activity
 GO:00198340.00803713Phospholipase A2 inhibitor activity
 GO:00037290.008574252mRNA binding
 GO:00050470.01070214Signal recognition particle binding
SVTE
 GO:0003735 4.69×10−2012158Structural constituent of ribosome
 GO:0003723 2.79×10−56630RNA binding
 GO:00152660.00124511Protein channel activity
 GO:00037290.001876252mRNA binding
 GO:00150770.0077022107Monovalent inorganic cation transmembrane transporter activity
 GO:00081210.00991718 Ubiquinol-cytochrome C reductase activity
 GO:00166790.00991718Oxidoreductase activity, acting on diphenols and associated substances as donors
 GO:00305520.017294114cAMP binding
 GO:00050800.018518115Protein kinase C binding
 GO:00086030.019741116cAMP-dependent protein kinase regulator activity
Non-overlap
 GO:0003735 2.55×10−65158Structural constituent of ribosome
 GO:00055200.000583225Insulin-like growth factor binding
 GO:00476960.00288112Beta-adrenergic receptor kinase activity
 GO:00050100.00431813Insulin-like growth factor receptor activity
 GO:00198340.00431813Phospholipase A2 inhibitor activity
 GO:00050470.00575314Signal recognition particle binding
 GO:00350350.00861816Histone acetyltransferase binding
 GO:00083120.008618167S RNA binding
 GO:00047030.01004817G-protein coupled receptor kinase activity
 GO:00313690.01147518Translation initiation factor binding

[i] GO, gene ontology; DEG, differentially expressed genes; RVTE, recurrent venous thromboembolism; SVTE, single venous thromboembolism; Non-overlap, non-overlapping DEGs as specific DEGs of RVTE; cAMP, cyclic adenosine monophosphate; GOMFID, gene ontology molecular function identification number ; Count, DEG numbers enriched in the specific GO term; Size, total gene numbers in the GO database.

In SVTE, the most significantly enriched GO terms for DEGs in biological processes included biopolymer biosynthesis (P=2.35×10−5), translational elongation (P=2.35×10−5), cellular protein metabolism (P=1.24×10−7) and rRNA processing (P=0.006). The predominantly enriched GO terms of the cellular component category were the cytosol (P=1.68×10−11), the cytosolic large ribosomal sub-unit (P=8.62×10−10) and macromolecular complexes (P=4.86×10−9). The main GO terms of DEGs in SVTE in the molecular function category were structural constituents of ribosomes (P=4.69×10−20), RNA binding (P=2.79×10−5) and protein channel activity (P=0.001). Among the DEGs in SVTE, 12 genes, including RPS3A and RPS7, were involved in translational elongation, 2 genes (UQCRQ and COX7C) were involved in oxidoreductase activity and 2 genes (HINT1 and LAMP1) were associated with proteolysis (Tables IIIV).

The GO terms for biological processes of the 22 specific DEGs of RVTE were mainly translational elongation (P=3.52×10−7), negative regulation of the force of heart contraction by chemical signals (P=0.001) and cell proliferation (P=0.002). In the cellular component category, enriched GO terms were mainly associated with the cytosolic small ribosomal sub-unit (P=1.56×10−5), ribosomal sub-unit (P=1.84×10−5) and macromolecular complexes (P=0.0008). The specific GO terms of the 22 specific DEGs in RVTE in the molecular function category were mainly associated with structural constituents of ribosomes (P=2.55×10−6), insulin-like growth factor binding (P=0.0006), beta-adrenergic receptor kinase activity (P=0.003) and phospholipase A2 inhibitor activity (P=0.004). Two genes (IGF2R and IGFBP7) were shown to be involved in anti-apoptotic signaling, ten genes, including RPL21 and RPS21, were involved in translational elongation and six genes, including NDUFA5, ATP5O and ADRBK1, were associated with the force of heart contraction (Tables IIIV).

Pathway enrichment analysis of DEGs

The DEGs identified in the present study were enriched in nine pathways (Table V). The RVTE DEGs were mainly enriched in ribosomal pathways (P=1.59×10−23), Parkinson's disease (P=0.007) and oxidative phosphorylation (P=0.008). The SVTE DEGs were enriched in ribosomal pathways (P=4.58×10−19) and cardiac muscle contraction (P=0.025). The non-overlapping DEGs were enriched in ribosomal pathways (P=2.25×10−6) and protein export (P=0.03).

Table V

KEGG pathway enrichment analysis of DEGs in patients with RVTE and SVTE, and specific DEGs of RVTE.

Table V

KEGG pathway enrichment analysis of DEGs in patients with RVTE and SVTE, and specific DEGs of RVTE.

Category/KEGGIDP-valueCount (n)Size (n)Term
RVTE
 K03010 1.59×10−231786Ribosome
 K050120.0073774133Parkinson's disease
 K001900.0077724135Oxidative phosphorylation
 K050100.0174214171Alzheimer's disease
 K050160.0225944185Huntington's disease
SVTE
 K03010 4.58×10−191286Ribosome
 K042600.025106279Cardiac muscle contraction
Non-overlap
 K03010 2.25×10−6586Ribosome
 K030600.03007111Protein export

[i] KEGG, Kyoto Encyclopedia of Genes and Genomes; DEG, differentially expressed genes; RVTE, recurrent venous thromboembolism; SVTE, single venous thromboembolism; Non-overlap, non-overlapping DEGs as specific DEGs of RVTE. KEGGID, KEGG identification number. Count, number of DEGs enriched in the specific GO term; size, number of genes in the GO database.

Metabolic pathway visualization analysis of specific DEGs of RVTE

The metabolic pathways were visualized in schemes depicting the ribosomal pathway (Fig. 2) and the protein export pathway (Fig. 3; http://www.genome.jp/kegg/tool/map_pathway2.html). In the ribosomal pathway, RPL21, RPS21, RPS24 and RPS27 were upregulated and in the protein export pathway, SRP9 was upregulated. The results suggested that these genes may be critical in RVTE and that certain variations in the expression of these genes may lead to an increased risk of recurrence.

Discussion

In recent years, the application of adequate thromboprophylaxis has led to significant progress in the management of VTE by successfully reducing morbidity and mortality (21). However, to date, methods for effectively preventing and diagnosing SVTE and RVTE have remained controversial (22). The present study used bioinformatics methods to investigate the molecular mechanisms and potential biomarkers of SVTE and RVTE.

In the present study, gene expression profiles of whole blood samples were successfully used to screen for DEGs in specimens from patients with SVTE compared with those in control specimens. With regard to enriched biological processes and pathways for DEGs in SVTE, genes involved in ribosomal pathways, including RPS3A and RPS7, and mitochondrial function, including UQCRQ and COX7C, were indicated to be most consistently affected and modulated. Ribosomal proteins have remained highly conserved during evolution and reflect critical functions in ribosome biogenesis; in addition, several ribosomal proteins were shown to have extra-ribosomal functions in apoptosis, DNA repair and genetic disease (23). A total of 12 DEGs were involved in ribosomal pathways. A paucity of studies have explored the pathogenesis of VTE. It has previously been indicated that the ribosomal-related RP-MDM2-P53 axis may be involved in the molecular pathogenesis of the 5q syndrome, and VTE was reported in 3% of patients with 5q syndrome (24). COX7C and UQCRQ are constituents of the mitochondrial respiratory chain (25). Mutations of these two genes may increase oxidative stress in coronary artery disease (26). The mortality after VTE is strongly associated with presentation of underlying cardiovascular disease (27). First-time VTE in numerous patients is idiopathic and challenging to diagnose, while COX7C and UQCRQ may represent novel biomarkers to identify SVTE.

The specific DEGs in RVTE were found to be mainly involved in ribosomal pathways, heart contraction and oxidative phosphorylation. Pathway visualization revealed that RPL21, RPS21, RPS24 and RPS27, which all encode ribosomal proteins, were enriched in ribosomal pathways, while SRP9 was enriched in the protein export pathway. Furthermore, RPL21, RPS21, RPS24 and RPS27 were found to be involved in RVTE through critical ribosome biogenesis or extra-ribosomal functions; of note, the expression of these genes was upregulated in patients with RVTE but unchanged in patients with SVTE. It has been reported that certain diseases, including transient ischemia/reperfusion and preeclampsia, are associated with ribosomes (28,29). RPS24 mutation was potentially linked to pathologies of Diamond-Blackfan anemia (30). The above results suggested that RPL21, RPS21, RPS24 and RPS27 may have critical roles in RVTE.

The results of the present study showed that β-adrenergic receptor kinase 1 (ADRBK1) was closely associated with RVTE and involved in heart contraction. Polymorphisms in ADRB2 and LPL genes are known to have central roles in vascular biology (31). A previous study suggested that the use of ADRBK1 as a biomarker significantly improved heart failure therapy (32). Certain studies showed that the ADRBK1/phosphoinositide-3 kinase (PI3K) complex improved cardiac function and that PI3K-dependent phosphorylation of ADRBK1 on Ser670 is responsible for the downregulation of ADRBK1 protein via a proteasome-dependent pathway (33,34). Furthermore, PI3K influences insulin-like growth factor and blood vessel-related factor through G protein beta gamma (35). These results may suggest that ADRBK1 has a critical role in RVTE by serving as a dual effector of the compensatory myocardial diastole and the PI3K response.

The present study also identified NDUFA5 and ATP ATP5O as DEGs, which were significantly associated with oxidative phosphorylation. The oxidative stress injury and exitotoxicity in mitochondria induced by NDUFA5 and ATP5O have been proved to be the cause of a variety of nervous system degenerative diseases, including Parkinson's, Alzheimer's and Huntington's disease (36,37). Free-radical generation and consequent oxidative stress in platelet activation and thrombotic vascular diseases have a distinctive role with the potential injurious effects of homocysteine (38,39). Therefore, NDUFA5 and ATP5O inducing oxidative stress injury and exitotoxicity in mitochondria may also have an impact on RVTE.

In conclusion, the screening performed in the present study identified 42 DEGs in RVTE, including 35 up- and 7 downregulated genes, 20 DEGs in SVTE, including 17 up- and 3 downregulated genes, and 22 specific DEGs in RVTE. Furthermore, functional and pathway enrichment analysis was performed for these identified DEGs. The results indicated that DEGs in SVTE, including COX7C and UQCRQ, may be used as potential biomarkers for SVTE and that specific DEGs in RVTE, including ADRBK1, NDUFA5 and ATP5O, may be considered as potential biomarkers of RVTE. However, experimental studies are required to confirm these results.

Acknowledgments

The present study was supported by the National Natural Science Foundation of China (grant no. 81370255).

References

1 

Pabinger I and Ay C: Biomarkers and venous thromboembolism. Arterioscler Thromb Vasc Biol. 29:332–336. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Baglin T: What happens after venous thromboembolism? J Thromb Haemost. 7(Suppl 1): 287–290. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Stein PD, Beemath A and Olson RE: Trends in the incidence of pulmonary embolism and deep venous thrombosis in hospitalized patients. Am J Cardiol. 95:1525–1526. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Nielsen JD: The incidence of pulmonary embolism during deep vein thrombosis. Phlebology. 28(Suppl 1): S29–S33. 2013. View Article : Google Scholar

5 

Heit JA, Mohr DN, Silverstein MD, Petterson TM, O'Fallon WM and Melton LJ III: Predictors of recurrence after deep vein thrombosis and pulmonary embolism: A population-based cohort study. Arch Intern Med. 160:761–768. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Poli D and Palareti G: Assessing recurrence risk following acute venous thromboembolism: Use of algorithms. Curr Opin Pulm Med. 19:407–412. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Barnes DM, Wakefield TW and Rectenwald JE: Novel biomarkers associated with deep venous thrombosis: A comprehensive review. Biomarker Insights. 3:93–100. 2008.PubMed/NCBI

8 

Kyrle PA, Minar E, Hirschl M, Bialonczyk C, Stain M, Schneider B, Weltermann A, Speiser W, Lechner K and Eichinger S: High plasma levels of factor VIII and the risk of recurrent venous thromboembolism. N Engl J Med. 343:457–462. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Comp PC and Esmon CT: Recurrent venous thromboembolism in patients with a partial deficiency of protein S. N Engl J Med. 311:1525–1528. 1984. View Article : Google Scholar : PubMed/NCBI

10 

Palareti G, Cosmi B, Legnani C, Tosetto A, Brusi C, Iorio A, Pengo V, Ghirarduzzi A, Pattacini C, Testa S, et al: D-dimer testing to determine the duration of anticoagulation therapy. N Engl J Med. 355:1780–1789. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Pabinger I, Thaler J and Ay C: Biomarkers for prediction of venous thromboembolism in cancer. Blood. 122:2011–2018. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Ay C, Dunkler D, Simanek R, Thaler J, Koder S, Marosi C, Zielinski C and Pabinger I: Prediction of venous thromboembolism in patients with cancer by measuring thrombin generation: Results from the Vienna cancer and thrombosis study. J Clin Oncol. 29:2099–2103. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Bouman AC, Smits JJ, Ten Cate H and Ten Cate-Hoek AJ: Markers of coagulation, fibrinolysis and inflammation in relation to post-thrombotic syndrome. J Thromb Haemost. 10:1532–1538. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Lewis DA, Stashenko GJ, Akay OM, Price LI, Owzar K, Ginsburg GS, Chi JT and Ortel TL: Whole blood gene expression analyses in patients with single vs. recurrent venous thromboembolism. Thromb Res. 128:536–540. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Gentleman RC, Carey VJ, Bates DM, Bolstad B, Dettling M, Dudoit S, Ellis B, Gautier L, Ge Y, Gentry J, et al: Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5:R802004. View Article : Google Scholar : PubMed/NCBI

16 

Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B and Speed TP: Summaries of affymetrix genechip probe level data. Nucleic Acids Res. 31:e152003. View Article : Google Scholar : PubMed/NCBI

17 

Wilson CL and Miller CJ: Simpleaffy: A Bioconductor package for affymetrix quality control and data analysis. Bioinformatics. 21:3683–3685. 2005. View Article : Google Scholar : PubMed/NCBI

18 

R Development Core Team: R: A Language and Environment for Statistical Computing. The R Foundation for Statistical Computing; Vienna, Austria: 2011, ISBN: 3-900051-07-0

19 

Zheng G, Wang H, Wei C and Li Y: iGepros: An integrated gene and protein annotation server for biological nature exploration. BMC Bioinformatics. 12(Suppl 14): S62011. View Article : Google Scholar

20 

Luo W and Brouwer C: Pathview: an R/Bioconductor package for pathway-based data integration and visualization. Bioinformatics. 29:1830–1831. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Verhamme P and Bounameaux H: Direct oral anticoagulants for acute venous thromboembolism: Closing the circle? Circulation. 129:725–727. 2014. View Article : Google Scholar

22 

Lippi G, Cervellin G, Franchini M and Favaloro EJ: Biochemical markers for the diagnosis of venous thromboembolism: The past, present and future. J Thromb Thrombolysis. 30:459–471. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Lindström MS: Emerging functions of ribosomal proteins in gene-specific transcription and translation. Biochem Biophys Res Commun. 379:167–170. 2009. View Article : Google Scholar

24 

Komrokji RS, Padron E, Ebert BL and List AF: Deletion 5q MDS: Molecular and therapeutic implications. Best Pract Res Clin Haematol. 26:365–375. 2013. View Article : Google Scholar

25 

Beech RD, Lowthert L, Leffert JJ, Mason PN, Taylor MM, Umlauf S, Lin A, Lee JY, Maloney K, Muralidharan A, et al: Increased peripheral blood expression of electron transport chain genes in bipolar depression. Bipolar disorders. 12:813–824. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Taurino C, Miller WH, McBride MW, McClure JD, Khanin R, Moreno MU, Dymott JA, Delles C and Dominiczak AF: Gene expression profiling in whole blood of patients with coronary artery disease. Clin Sci (Lond). 119:335–343. 2010. View Article : Google Scholar

27 

White RH: The epidemiology of venous thromboembolism. Circulation. 107:I4–I8. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Textoris J, Ivorra D, Ben Amara A, Sabatier F, Ménard JP, Heckenroth H, Bretelle F and Mege JL: Evaluation of current and new biomarkers in severe preeclampsia: A microarray approach reveals thevs.ig4 gene as a potential blood biomarker. PLoS One. 8:e826382013. View Article : Google Scholar

29 

White BC, Sullivan JM, DeGracia DJ, O'Neil BJ, Neumar RW, Grossman LI, Rafols JA and Krause GS: Brain ischemia and reperfusion: Molecular mechanisms of neuronal injury. J Neurol Sci. 179:1–33. 2000. View Article : Google Scholar : PubMed/NCBI

30 

Choesmel V, Fribourg S, Aguissa-Touré AH, Pinaud N, Legrand P, Gazda HT and Gleizes PE: Mutation of ribosomal protein RPS24 in Diamond-Blackfan anemia results in a ribosome biogenesis disorder. Hum Mol Genet. 17:1253–1263. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Zee RY, Cook NR, Cheng S, Erlich HA, Lindpaintner K and Ridker PM: Polymorphism in the beta2-adrenergic receptor and lipoprotein lipase genes as risk determinants for idiopathic venous thromboembolism: A multilocus, population-based, prospective genetic analysis. Circulation. 113:2193–2200. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Tachibana H, Naga Prasad SV, Lefkowitz RJ, Koch WJ and Rockman HA: Level of beta-adrenergic receptor kinase 1 inhibition determines degree of cardiac dysfunction after chronic pressure overload-induced heart failure. Circulation. 111:591–597. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Ito K, Akazawa H, Tamagawa M, Furukawa K, Ogawa W, Yasuda N, Kudo Y, Liao CH, Yamamoto R, Sato T, et al: PDK1 coordinates survival pathways and beta-adrenergic response in the heart. Proc Natl Acad Sci USA. 106:8689–8694. 2009. View Article : Google Scholar : PubMed/NCBI

34 

Lombardi MS, Vroon A, Sodaar P, van Muiswinkel FL, Heijnen CJ and Kavelaars A: Down-regulation of GRK2 after oxygen and glucose deprivation in rat hippocampal slices: Role of the PI3-kinase pathway. J Neurochem. 102:731–740. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Hideshima T, Nakamura N, Chauhan D and Anderson KC: Biologic sequelae of interleukin-6 induced PI3-K/Akt signaling in multiple myeloma. Oncogene. 20:5991–6000. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Chou JL, Shenoy DV, Thomas N, Choudhary PK, Laferla FM, Goodman SR and Breen GA: Early dysregulation of the mitochondrial proteome in a mouse model of Alzheimer's disease. J Proteomics. 74:466–479. 2011. View Article : Google Scholar : PubMed/NCBI

37 

Weydt P, Pineda VV, Torrence AE, Libby RT, Satterfield TF, Lazarowski ER, Gilbert ML, Morton GJ, Bammler TK, Strand AD, et al: Thermoregulatory and metabolic defects in Huntington's disease transgenic mice implicate PGC-1alpha in Huntington's disease neurodegeneration. Cell Metab. 4:349–362. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Davi G, Di Minno G, Coppola A, Andria G, Cerbone AM, Madonna P, Tufano A, Falco A, Marchesani P, Ciabattoni G and Patrono C: Oxidative stress and platelet activation in homozygous homocystinuria. Circulation. 104:1124–1128. 2001. View Article : Google Scholar : PubMed/NCBI

39 

El Kossi MM and Zakhary MM: Oxidative stress in the context of acute cerebrovascular stroke. Stroke. 31:1889–1892. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2015
Volume 12 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhou W, Zhang K, Chen D, Gao P and Wang Q: Gene microarray analyses for potential biomarkers of single and recurrent venous thromboembolism. Mol Med Rep 12: 7358-7366, 2015
APA
Zhou, W., Zhang, K., Chen, D., Gao, P., & Wang, Q. (2015). Gene microarray analyses for potential biomarkers of single and recurrent venous thromboembolism. Molecular Medicine Reports, 12, 7358-7366. https://doi.org/10.3892/mmr.2015.4349
MLA
Zhou, W., Zhang, K., Chen, D., Gao, P., Wang, Q."Gene microarray analyses for potential biomarkers of single and recurrent venous thromboembolism". Molecular Medicine Reports 12.5 (2015): 7358-7366.
Chicago
Zhou, W., Zhang, K., Chen, D., Gao, P., Wang, Q."Gene microarray analyses for potential biomarkers of single and recurrent venous thromboembolism". Molecular Medicine Reports 12, no. 5 (2015): 7358-7366. https://doi.org/10.3892/mmr.2015.4349