Skip to main content
Erschienen in: European Journal of Medical Research 1/2014

Open Access 01.12.2014 | Research

Construction of microRNA and transcription factor regulatory network based on gene expression data in cardiomyopathy

verfasst von: Lei Wang, Jihua Hu, Haijian Xing, Min Sun, Juanli Wang, Qiang Jian, Hua Yang

Erschienen in: European Journal of Medical Research | Ausgabe 1/2014

Abstract

Background

Cardiomyopathy is a progressive myocardial disorder. Here, we attempted to reveal the possible mechanism of cardiomyopathy at the transcription level with the roles of microRNAs (miRNAs) and transcription factors (TFs) taken into account.

Method

We firstly identified differentially expressed genes (DEGs) between cardiomyopathy patients and controls with data from the gene expression omnibus (GEO) database. DEGs were associated with the canonical pathways, molecular and cellular functions, physiological system development and function in the Ingenuity Knowledge Base by using the Ingenuity Pathway Analysis (IPA) software. TFs and miRNAs that DEGs significantly enriched were identified and a double-factor regulatory network was constructed.

Results

A total of 1,680 DEGs were identified. The DEGs were enriched for various pathways, with glucocorticoid receptor signaling as the most significant. A double-factor regulatory network was constructed, including seven TFs and two miRNAs. A subnetwork under the regulation of MEF2C and SRF was also constructed to illustrate their regulatory effects on cardiac functions.

Conclusion

Our results may provide new understanding of cardiomyopathy and may facilitate further therapeutic studies.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s40001-014-0057-5) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.
Abkürzungen
DEG
differentially expressed genes
GEO
Gene Expression Omnibus
IPA
Ingenuity Pathway Analysis
miRNAs
microRNAs
RMA
Robust Multi-array Analysis
TF
transcription factor
UCSC
University of California Santa Cruz

Background

Cardiomyopathy is a progressive myocardial disorder, usually leading to cardiovascular death or heart failure-related disability [1]. It develops at any age, in either sex, and in any population [2],[3]. The etiology of cardiomyopathy is highly complex and improving its treatments has become a research hotspot.
Using bioinformatics combined with gene expression data to identify potential therapeutic targets has shown great application prospects. The majority of the previous studies mainly focused on the analyses of differentially expressed genes (DEGs), without considering microRNAs (miRNAs) and transcription factors (TFs) that regulate the expression of DEGs. miRNAs are small non-coding RNAs that control various biological processes through affecting the stability and translation of target mRNAs. Previous studies have proposed several miRNAs as being involved in the pathogenesis of cardiomyopathy, such as miR-1 [4] and miR-21 [5]. TFs can regulate gene expression through binding to the cis-elements in target genes' promoter regions. TFs, such as MEF2, have been reported to be associated with cardiomyopathy [6]. Considering the important regulatory roles of miRNAs and TFs in the pathogenesis of cardiomyopathy, identification of miRNAs and TFs that enriched with target DEGs and construction of a double-factor regulatory network may provide new understanding of the molecular mechanism of cardiomyopathy.
In the current study, based on gene expression data from the Gene Expression Omnibus (GEO) database, we acquired DEGs, miRNAs and TFs that enriched with target DEGs and constructed a double-factor regulatory network. Our results may reveal the possible mechanism of cardiomyopathy at the transcription level.

Methods

Ethics Statement

This study was approved by the institutional review board of the Xi'an Children's Hospital (20140518). Written informed consent was obtained from all patients for the publication of this report and any accompanying images.

Microarray data

The gene expression profile GSE5406 from the GEO database was used. This dataset includes transcription profiles of 210 left ventricular myocardial tissue samples, 194 of which were from patients with advanced cardiomyopathy and 16 of which were from healthy donors. All patients had New York Heart Association class 3 to 4 symptoms and left ventricular systolic dysfunction, with ejection fraction of 14 ± 8% (mean ± SD). Patients suffered from heart failure due to ischemic (n =86) or idiopathic dilated (n =108) cardiomyopathy. Control samples had normal left ventricular function with ejection fraction of 56 ± 7% (P =0.0001 versus patients). None of the subjects received mechanical support with left ventricular assist devices. Myocardial tissue samples were obtained from patients undergoing heart transplantation and from controls deemed unsuitable for transplantation. Whole hearts were removed after preservation in cold cardioplegia at the time of transplantation or donor harvest. Then, segments of noninfarcted left ventricular free wall were snap-frozen in liquid nitrogen. For each sample, RNA was isolated by using Trizol reagent (Invitrogen, Carlsbad, CA, USA). The dataset was generated by using the (HG-U133A) Affymetrix Human Genome U133A Array (Affymetrix, Santa Clara, CA, USA).

Identification of differentially expressed genes (DEGs)

Raw data from all arrays were normalized using Robust Multi-array Analysis (RMA) [7] in the R software (version 3.0.0). The resulting expression values were used to identify DEGs with the limma package (3.12.1) in R. DEGs were detected by using t-tests and multiple test corrections were carried out with the Benjamini-Hochberg method [8]. The threshold for significance was set as P <0.01.

Enrichment analysis

To explore the functions and pathways of DEGs, DEGs were associated with the canonical pathways, molecular and cellular functions, physiological system development and function in the Ingenuity Knowledge Base by using the Ingenuity Pathway Analysis (IPA) software (Ingenuity® Systems, http://​www.​ingenuity.​com).

Construction of miRNA-TF regulatory network

We also acquired miRNAs and TFs which were overrepresented with target DEGs. For miRNA analysis, we used Targetscan [9], miRanda [10] and Pita [11] for miRNA target DEGs prediction. To avoid false positive results, miRNA-DEG prediction results supported by all three prediction methods were considered to be confidential. For TF analysis, we obtained TF binding sites and the coordinate's position information of human being from the University of California Santa Cruz (UCSC) database. TFs and miRNAs that DEGs significantly enriched were identified by using the hypergeometric distribution test. P <0.01 was set as the threshold. Based on the regulatory relationship between miRNAs or TFs and DEGs, a network was constructed by using the IPA software.

Results

A total of 1,680 DEGs were identified, including 963 down-regulated genes and 717 up-regulated ones in advanced cardiomyopathy patients.
As shown in Figure 1, the IPA results of canonical pathways showed that the DEGs were enriched for various pathways, among which glucocorticoid receptor signaling was the most significant. Molecular and cellular functions analysis revealed that `Cell Death and Survival' (P =2.53 × 10–24 to 1.36 × 10–3) was the top molecular function affected by DEGs followed by `Cellular Growth and Proliferation' (P =3.37 × 10–24 to 1.36 × 10–3) (Table 1). `Cardiovascular System Development and Function' (P =4.17 × 10–12 to 1.20 × 10–3) was the top physiological function mediated by DEGs (Table 1).
Table 1
Ingenuity Pathway Analysis: functions related to differentially expressed genes
Name
P-value
Number of molecules
Molecular and Cellular Functions
Cell Death and Survival
2.53 × 10–24 to 1.36 × 10–3
405
Cellular Growth and Proliferation
3.37 × 10–24 to 1.36 × 10–3
414
Cellular Movement
5.41 × 10–13 to 1.36 × 10–3
236
Gene Expression
8.14 × 10–11 to 1.20 × 10–3
247
Cellular Development
1.13 × 10–10 to 1.36 × 10–3
328
Physiological System Development and Function
Cardiovascular System Development and Function
4.17 × 10–12 to 1.20 × 10–3
125
Organismal Development
3.96 × 10–11 to 1.22 × 10–3
92
Tissue Development
3.68 × 10–8 to 1.22 × 10–3
163
Tumor Morphology
3.84 × 10–7 to 1.20 × 10–3
117
Connective Tissue Development and Function
8.52 × 10–6 to 1.24 × 10–3
40
The results of miRNA and TF enrichment analysis are listed in Table 2. A network was constructed to illustrate the regulatory relationships (Figure 2). Considering the important role of SRF and MEF2C in the progression of cardiomyopathy, a subnetwork under their regulation was constructed and the biological functions they may affect were also indicated (Figure 3).
Table 2
Transcription factors and microRNAs (miRNAs) enriched with target differentially expressed genes (DEGs)
Name
P-value
MicroRNA
miR-30c-5p (and other miRNAs w/seed GUAAACA)
3.12 × 10–4
miR-125b-5p (and other miRNAs w/seed CCCUGAG)
2.15 × 10–3
Transcription factors
MEF2C
5.65 × 10–6
GATA1
6.11 × 10–5
SRF
1.53 × 10–4
NUPR1
4.18 × 10–4
ATF4
7.25 × 10–4
RELA
9.35 × 10–3
GLI2
1.42 × 10–2

Discussion

In the current study, based on GSE5406 from the GEO database, 1,680 DEGs were identified between advanced cardiomyopathy patients and healthy controls. DEGs were mapped into the Ingenuity Knowledge Base and several canonical pathways were screened out, with glucocorticoid receptor signaling as the most significant. Glucocorticoid receptors are involved in cardiovascular homeostasis [12] and glucocorticoid could protect rodent hearts from ischemia/reperfusion injury [13]. Our results confirmed the essential roles of this pathway in the pathogenesis of cardiomyopathy. Molecular and cellular functions analysis revealed that `Cell Death and Survival' (P =2.53 × 10–24 to 1.36 × 10–3) was the top molecular function affected by DEGs, suggesting that dysregulation of the cell function may contribute to the progressive heart failure induced by cardiomyopathy.
TFs or miRNAs that DEGs significantly enriched were identified and a double-factor regulatory network was constructed, including seven TFs and two miRNAs (Figure 2). All these miRNAs and TFs have previously been implicated in cardiomyogenesis or cardiac function. Considering their important regulatory roles in controlling various biological processes, these miRNAs and TFs may provide new avenues for the therapeutic strategies of cardiomyopathy. In addition, since all these miRNAs and TFs were detected in patients suffered from New York Heart Association class 3 to 4 symptoms, they may also be considered as prognosis markers in clinical practice. A subnetwork under the regulation of SRF and MEF2C was also constructed and the functions they may affect were also indicated (Figure 3) considering the considerable evidence in support of their involvement in cardiomyopathy [14]-[17]. As shown in Figure 3, both SRF and MEF2C may inhibit the expression of MYH6 and CNN1. MYH6 is a famous cardiac muscle myosin gene and mutations of this gene are implicated in cardiomyopathy in various studies [18]. Inhibition of CNN1 expression may disrupt its suppression of cardiomyopathy through the εPKC pathway [19]. In addition, both SRF and MEF2C may regulate the expression of NPPA, which was overexpressed in patients in our results. This gene is involved in many cardiac dysfunctions (Figure 3), including myocardial infarction, heart failure, and high ventricular pressure [20],[21].

Conclusion

In summary, with DEGs identified by gene chip data generated by high-throughput technologies from the GEO database, we constructed a cardiomyopathy double-factor regulatory network and revealed the possible mechanism on regulation level. A subnetwork under the regulation of SRF and MEF2C was also constructed to illustrate their possible regulatory roles. Our results may facilitate further therapeutic studies of cardiomyopathy.

Authors' contributions

LW conceived of the study and drafted the manuscript. JH, HX and MS performed the statistical analysis. JW, QJ and HY revised the manuscript. All authors read and approved the final manuscript.

Acknowledgement

This study is supported by Provincial Natural Science Basic Research Foundation of Shaanxi (2014JM4152) and the Science and Technology Project of Xi'an Municipal Health Bureau (Number 2013028).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.
Anhänge

Authors’ original submitted files for images

Literatur
1.
Zurück zum Zitat Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D, Moss AJ, Seidman CE, Young JB: Contemporary definitions and classification of the cardiomyopathies: an American Heart Association scientific statement from the council on clinical cardiology, heart failure and transplantation committee; quality of care and outcomes research and functional genomics and translational biology interdisciplinary working groups; and council on epidemiology and prevention. Circulation 2006, 113: 1807–1816. 10.1161/CIRCULATIONAHA.106.174287PubMedCrossRef Maron BJ, Towbin JA, Thiene G, Antzelevitch C, Corrado D, Arnett D, Moss AJ, Seidman CE, Young JB: Contemporary definitions and classification of the cardiomyopathies: an American Heart Association scientific statement from the council on clinical cardiology, heart failure and transplantation committee; quality of care and outcomes research and functional genomics and translational biology interdisciplinary working groups; and council on epidemiology and prevention. Circulation 2006, 113: 1807–1816. 10.1161/CIRCULATIONAHA.106.174287PubMedCrossRef
2.
Zurück zum Zitat Towbin JA, Lowe AM, Colan SD, Sleeper LA, Orav EJ, Clunie S, Messere J, Cox GF, Lurie PR, Hsu D, Canter C, Wilkinson JD, Lipshultz SE: Incidence, causes, and outcomes of dilated cardiomyopathy in children. JAMA 2006, 296: 1867–1876. 10.1001/jama.296.15.1867PubMedCrossRef Towbin JA, Lowe AM, Colan SD, Sleeper LA, Orav EJ, Clunie S, Messere J, Cox GF, Lurie PR, Hsu D, Canter C, Wilkinson JD, Lipshultz SE: Incidence, causes, and outcomes of dilated cardiomyopathy in children. JAMA 2006, 296: 1867–1876. 10.1001/jama.296.15.1867PubMedCrossRef
3.
Zurück zum Zitat Taylor MR, Carniel E, Mestroni L: Cardiomyopathy, familial dilated. Orphanet J Rxare Dis 2006, 1: 27. 10.1186/1750-1172-1-27CrossRef Taylor MR, Carniel E, Mestroni L: Cardiomyopathy, familial dilated. Orphanet J Rxare Dis 2006, 1: 27. 10.1186/1750-1172-1-27CrossRef
4.
Zurück zum Zitat Ferreira LR, Frade AF, Santos RH, Teixeira PC, Baron MA, Navarro IC, Benvenuti LA, Fiorelli AI, Bocchi EA, Stolf NA, Chevillard C, Kalil J, Cunha-Neto E: MicroRNAs miR-1, miR-133a, miR-133b, miR-208a and miR-208b are dysregulated in chronic Chagas disease cardiomyopathy. Int J Cardiol 2014, 175: 409–417. 10.1016/j.ijcard.2014.05.019PubMedCrossRef Ferreira LR, Frade AF, Santos RH, Teixeira PC, Baron MA, Navarro IC, Benvenuti LA, Fiorelli AI, Bocchi EA, Stolf NA, Chevillard C, Kalil J, Cunha-Neto E: MicroRNAs miR-1, miR-133a, miR-133b, miR-208a and miR-208b are dysregulated in chronic Chagas disease cardiomyopathy. Int J Cardiol 2014, 175: 409–417. 10.1016/j.ijcard.2014.05.019PubMedCrossRef
5.
Zurück zum Zitat Xu HF, Ding YJ, Zhang ZX, Wang ZF, Luo CL, Li BX, Shen YW, Tao LY, Zhao ZQ: MicroRNA21 regulation of the progression of viral myocarditis to dilated cardiomyopathy. Mol Med Reports 2014, 10: 161–168. Xu HF, Ding YJ, Zhang ZX, Wang ZF, Luo CL, Li BX, Shen YW, Tao LY, Zhao ZQ: MicroRNA21 regulation of the progression of viral myocarditis to dilated cardiomyopathy. Mol Med Reports 2014, 10: 161–168.
6.
Zurück zum Zitat Hannenhalli S, Putt ME, Gilmore JM, Wang J, Parmacek MS, Epstein JA, Morrisey EE, Margulies KB, Cappola TP: Transcriptional genomics associates FOX transcription factors with human heart failure. Circulation 2006, 114: 1269–1276. 10.1161/CIRCULATIONAHA.106.632430PubMedCrossRef Hannenhalli S, Putt ME, Gilmore JM, Wang J, Parmacek MS, Epstein JA, Morrisey EE, Margulies KB, Cappola TP: Transcriptional genomics associates FOX transcription factors with human heart failure. Circulation 2006, 114: 1269–1276. 10.1161/CIRCULATIONAHA.106.632430PubMedCrossRef
7.
Zurück zum Zitat Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 2003, 4: 249–264. 10.1093/biostatistics/4.2.249PubMedCrossRef Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U, Speed TP: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 2003, 4: 249–264. 10.1093/biostatistics/4.2.249PubMedCrossRef
8.
Zurück zum Zitat Benjamini Y, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B Methodol 1995, 57: 289–300. Benjamini Y, Hochberg Y: Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Ser B Methodol 1995, 57: 289–300.
9.
Zurück zum Zitat Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP: MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell 2007, 27: 91–105. 10.1016/j.molcel.2007.06.017PubMedPubMedCentralCrossRef Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP: MicroRNA targeting specificity in mammals: determinants beyond seed pairing. Mol Cell 2007, 27: 91–105. 10.1016/j.molcel.2007.06.017PubMedPubMedCentralCrossRef
10.
11.
Zurück zum Zitat Kertesz M, Iovino N, Unnerstall U, Gaul U, Segal E: The role of site accessibility in microRNA target recognition. Nat Genet 2007, 39: 1278–1284. 10.1038/ng2135PubMedCrossRef Kertesz M, Iovino N, Unnerstall U, Gaul U, Segal E: The role of site accessibility in microRNA target recognition. Nat Genet 2007, 39: 1278–1284. 10.1038/ng2135PubMedCrossRef
12.
Zurück zum Zitat Rickard AJ, Funder JW, Morgan J, Fuller PJ, Young MJ: Does glucocorticoid receptor blockade exacerbate tissue damage after mineralocorticoid/salt administration? Endocrinology 2007, 148: 4829–4835. 10.1210/en.2007-0209PubMedCrossRef Rickard AJ, Funder JW, Morgan J, Fuller PJ, Young MJ: Does glucocorticoid receptor blockade exacerbate tissue damage after mineralocorticoid/salt administration? Endocrinology 2007, 148: 4829–4835. 10.1210/en.2007-0209PubMedCrossRef
13.
Zurück zum Zitat Tokudome S, Sano M, Shinmura K, Matsuhashi T, Morizane S, Moriyama H, Tamaki K, Hayashida K, Nakanishi H, Yoshikawa N, Shimizu N, Endo J, Katayama T, Murata M, Yuasa S, Kaneda R, Tomita K, Eguchi N, Urade Y, Asano K, Utsunomiya Y, Suzuki T, Taguchi R, Tanaka H, Fukuda K: Glucocorticoid protects rodent hearts from ischemia/reperfusion injury by activating lipocalin-type prostaglandin D synthase-derived PGD2 biosynthesis. J Clin Invest 2009, 119: 1477–1488. 10.1172/JCI37413PubMedPubMedCentralCrossRef Tokudome S, Sano M, Shinmura K, Matsuhashi T, Morizane S, Moriyama H, Tamaki K, Hayashida K, Nakanishi H, Yoshikawa N, Shimizu N, Endo J, Katayama T, Murata M, Yuasa S, Kaneda R, Tomita K, Eguchi N, Urade Y, Asano K, Utsunomiya Y, Suzuki T, Taguchi R, Tanaka H, Fukuda K: Glucocorticoid protects rodent hearts from ischemia/reperfusion injury by activating lipocalin-type prostaglandin D synthase-derived PGD2 biosynthesis. J Clin Invest 2009, 119: 1477–1488. 10.1172/JCI37413PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Alonso-Montes C, Naves-Diaz M, Fernandez-Martin JL, Rodriguez-Reguero J, Moris C, Coto E, Cannata-Andia JB, Rodriguez I: New polymorphisms in human MEF2C gene as potential modifier of hypertrophic cardiomyopathy. Mol Biol Rep 2012, 39: 8777–8785. 10.1007/s11033-012-1740-7PubMedCrossRef Alonso-Montes C, Naves-Diaz M, Fernandez-Martin JL, Rodriguez-Reguero J, Moris C, Coto E, Cannata-Andia JB, Rodriguez I: New polymorphisms in human MEF2C gene as potential modifier of hypertrophic cardiomyopathy. Mol Biol Rep 2012, 39: 8777–8785. 10.1007/s11033-012-1740-7PubMedCrossRef
15.
Zurück zum Zitat Xu J, Gong NL, Bodi I, Aronow BJ, Backx PH, Molkentin JD: Myocyte enhancer factors 2A and 2C induce dilated cardiomyopathy in transgenic mice. J Biol Chem 2006, 281: 9152–9162. 10.1074/jbc.M510217200PubMedCrossRef Xu J, Gong NL, Bodi I, Aronow BJ, Backx PH, Molkentin JD: Myocyte enhancer factors 2A and 2C induce dilated cardiomyopathy in transgenic mice. J Biol Chem 2006, 281: 9152–9162. 10.1074/jbc.M510217200PubMedCrossRef
16.
Zurück zum Zitat Dasgupta T, Stillwagon SJ, Ladd AN: Gene expression analyses implicate an alternative splicing program in regulating contractile gene expression and serum response factor activity in mice. PLoS One 2013, 8: e56590. 10.1371/journal.pone.0056590PubMedPubMedCentralCrossRef Dasgupta T, Stillwagon SJ, Ladd AN: Gene expression analyses implicate an alternative splicing program in regulating contractile gene expression and serum response factor activity in mice. PLoS One 2013, 8: e56590. 10.1371/journal.pone.0056590PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Tritsch E, Mallat Y, Lefebvre F, Diguet N, Escoubet B, Blanc J, De Windt LJ, Catalucci D, Vandecasteele G, Li Z, Mericskay M: An SRF/miR-1 axis regulates NCX1 and annexin A5 protein levels in the normal and failing heart. Cardiovasc Res 2013, 98: 372–380. 10.1093/cvr/cvt042PubMedCrossRef Tritsch E, Mallat Y, Lefebvre F, Diguet N, Escoubet B, Blanc J, De Windt LJ, Catalucci D, Vandecasteele G, Li Z, Mericskay M: An SRF/miR-1 axis regulates NCX1 and annexin A5 protein levels in the normal and failing heart. Cardiovasc Res 2013, 98: 372–380. 10.1093/cvr/cvt042PubMedCrossRef
18.
Zurück zum Zitat Jiang J, Wakimoto H, Seidman JG, Seidman CE: Allele-specific silencing of mutant Myh6 transcripts in mice suppresses hypertrophic cardiomyopathy. Science 2013, 342: 111–114. 10.1126/science.1236921PubMedPubMedCentralCrossRef Jiang J, Wakimoto H, Seidman JG, Seidman CE: Allele-specific silencing of mutant Myh6 transcripts in mice suppresses hypertrophic cardiomyopathy. Science 2013, 342: 111–114. 10.1126/science.1236921PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Lu D, Zhang L, Bao D, Lu Y, Zhang X, Liu N, Ge W, Gao X, Li H, Zhang L: Calponin1 inhibits dilated cardiomyopathy development in mice through the epsilonPKC pathway. Int J Cardiol 2014, 173: 146–153. 10.1016/j.ijcard.2014.02.032PubMedCrossRef Lu D, Zhang L, Bao D, Lu Y, Zhang X, Liu N, Ge W, Gao X, Li H, Zhang L: Calponin1 inhibits dilated cardiomyopathy development in mice through the epsilonPKC pathway. Int J Cardiol 2014, 173: 146–153. 10.1016/j.ijcard.2014.02.032PubMedCrossRef
20.
Zurück zum Zitat Asanuma H, Kitakaze M: Carperitide and adiponectin - how are they connected each other to benefit acute decompensated heart failure? Circ J 2009, 73: 2206–2207. http://www.ncbi.nlm.nih.gov/pubmed/19940369 10.1253/circj.CJ-09-0785PubMedCrossRef Asanuma H, Kitakaze M: Carperitide and adiponectin - how are they connected each other to benefit acute decompensated heart failure? Circ J 2009, 73: 2206–2207. http://​www.​ncbi.​nlm.​nih.​gov/​pubmed/​19940369 10.1253/circj.CJ-09-0785PubMedCrossRef
21.
Zurück zum Zitat Tarazon E, Rosello-Lleti E, Rivera M, Ortega A, Molina-Navarro MM, Trivino JC, Lago F, Gonzalez-Juanatey JR, Orosa P, Montero JA, Salvador A, Portolés M: RNA sequencing analysis and atrial natriuretic peptide production in patients with dilated and ischemic cardiomyopathy. PLoS One 2014, 9: e90157. 10.1371/journal.pone.0090157PubMedPubMedCentralCrossRef Tarazon E, Rosello-Lleti E, Rivera M, Ortega A, Molina-Navarro MM, Trivino JC, Lago F, Gonzalez-Juanatey JR, Orosa P, Montero JA, Salvador A, Portolés M: RNA sequencing analysis and atrial natriuretic peptide production in patients with dilated and ischemic cardiomyopathy. PLoS One 2014, 9: e90157. 10.1371/journal.pone.0090157PubMedPubMedCentralCrossRef
Metadaten
Titel
Construction of microRNA and transcription factor regulatory network based on gene expression data in cardiomyopathy
verfasst von
Lei Wang
Jihua Hu
Haijian Xing
Min Sun
Juanli Wang
Qiang Jian
Hua Yang
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
European Journal of Medical Research / Ausgabe 1/2014
Elektronische ISSN: 2047-783X
DOI
https://doi.org/10.1186/s40001-014-0057-5

Weitere Artikel der Ausgabe 1/2014

European Journal of Medical Research 1/2014 Zur Ausgabe