Skip to main content
Erschienen in: Arthritis Research & Therapy 1/2015

Open Access 01.12.2015 | Research article

CD4 T-cell transcriptome analysis reveals aberrant regulation of STAT3 and Wnt signaling pathways in rheumatoid arthritis: evidence from a case–control study

verfasst von: Hua Ye, Jing Zhang, Jun Wang, Yanyan Gao, Yan Du, Chun Li, Minghua Deng, Jianping Guo, Zhanguo Li

Erschienen in: Arthritis Research & Therapy | Ausgabe 1/2015

Abstract

Introduction

Rheumatoid arthritis (RA) is a systemic autoimmune disease in which T cells play a pivotal role in the pathogenesis. Knowledge in terms of the CD4 T-cell transcriptome in RA is limited. The aim of this study was to examine the whole-genome transcription profile of CD4 T cells in RA by comparing patients with RA to healthy controls.

Methods

Peripheral blood CD4 T cells were isolated from 53 RA patients with active disease and 45 healthy individuals; 13 cases and 10 controls were enrolled in microarray analysis. The remaining 40 cases and 35 controls were recruited as an independent cohort for the validation study. Bioinformatics was performed on Gene Ontology (GO), gene-gene interaction networks, and pathway analysis. The gene modules, by combining the results from GO, gene networks, and pathway analysis, were selected for further validation.

Results

The CD4 T cells showed 1,496 differentially expressed (DE) genes in RA patients relative to healthy individuals. GO analysis revealed that the DE genes were enriched in immune response, T-cell response, apoptosis process, and Wnt receptor signaling. Pathway analysis also identified that ‘Wnt signaling pathway’ was differentially regulated between two groups (P = 2.78 × 10−10). By gene-gene network analysis, we found that the DE genes were enriched in T-cell receptor (TCR), JAK-STAT signaling, and Wnt signaling pathway. By gene module analysis, we found that a number of DE genes overlapped in the three different analyses. In total, 23 genes were selected for further validation, and nine genes were confirmed. Of these, four genes (SOCS3, CBL, IFNAR1, and PIK3CA) were involved in STAT3 (signal transducer and activator of transcription 3) signaling, and three genes (CBL, KLF9, and CSNK2A1) were involved in the Wnt signaling pathway. Additionally, several zinc finger transcription factors (ZEB1, ZNF292, and ZNF644) were confirmed.

Conclusions

We report here the first case–control study of the CD4 T-cell transcriptome profile in RA. Our data provide evidence that CD4 T cells from patients with RA have abnormal functional networks in STAT3 signaling and Wnt signaling. Our results also suggest that the aberrant expression of several zinc finger transcription factors (ZEB1, ZNF292, and ZNF644) may be potential pathogenic factors for RA.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13075-015-0590-9) contains supplementary material, which is available to authorized users.
Hua Ye and Jing Zhang contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HY contributed to the study design, clinical data collection, data interpretation, and manuscript drafting. JZ carried out real-time polymerase chain reaction experiments, participated in statistical analysis, and helped to revise the manuscript. JW helped to perform the bioinformatic analysis and revised the manuscript. YG, YD, and CL helped in the samples collection, clinical data interpretation, and manuscript revision. MD helped in the bioinformatic and statistical analysis and revised the manuscript. JG contributed to the study design and carried out the bioinformatic and statistical analysis, data interpretation, and manuscript drafting and revision. ZL conceived of the study, participated in its design, and helped to revise the manuscript. All authors read and approved the final manuscript.
Abkürzungen
DE
differentially expressed
FDR
false-discovery rate
FLS
fibroblast-like synoviocyte
GEO
Gene Expression Omnibus
GO
Gene Ontology
HPRD
Human Protein Reference Database
IFNγ
interferon-gamma
IL
interleukin
KEGG
Kyoto Encyclopedia of Genes and Genomes
MINT
Molecular INTeraction
MPEA
metabolite pathway enrichment analysis
NCBI
National Center for Biotechnology Information
PBMC
peripheral blood mononuclear cell
qPCR
quantitative real-time polymerase chain reaction
RA
rheumatoid arthritis
sFRP1
secreted frizzled-related protein 1
SLE
systemic lupus erythematosus
STAT3
signal transducer and activator of transcription 3
TCR
T-cell receptor
Th
T helper
Treg
regulatory T

Introduction

Rheumatoid arthritis (RA) is a chronic and systemic autoimmune disease characterized by symmetric inflammation in peripheral joints, leading to cartilage destruction and bone erosion. The etiology of RA remains elusive, but it is believed that genetic factors play an important role in RA pathogenesis. Multiple genes contribute to disease susceptibility and heterogeneous clinical manifestations [1,2]. To gain insight into the molecular signature underlying disease pathogenesis, gene expression profiling studies have emerged as a powerful way to comprehensively identify the genes that are differentially expressed in blood and tissues between patients and healthy individuals [3].
To date, a number of transcriptome studies have focused on peripheral blood mononuclear cells (PBMCs) or fibroblast-like synoviocytes (FLSs) to understand the aberrant biological pathways involved in the pathogenesis of RA [4-9]. Yet although much has been learned about T cells in RA pathology, knowledge is limited in terms of whole-genome transcription profiling of CD4 T cells in RA. One microarray study was conducted on CD4 T cells in RA but with a case-only design [10]. To better understand the complex molecular mechanisms and discover the potential predictive biomarkers for RA, we performed a case–control study of CD4 T-cell transcriptome analysis by comparing RA patients to healthy controls. We found a great difference in gene expression profiling of CD4 T cells between active RA cases and healthy controls and discovered several aberrant signaling pathways in CD4 T cells from patients with RA. Finally, by quantitative real-time polymerase chain reaction (qPCR) validation, we identified nine genes involved in STAT3 (signal transducer and activator of transcription 3) signaling, Wnt signaling pathway, and zinc finger transcription regulation.

Methods

Study subjects

Thirteen patients with RA and nine healthy controls were enrolled in microarray analysis. Forty RA cases and 35 healthy individuals were subsequently recruited for the validation study. All patients satisfied the American College of Rheumatology 1987 revised criteria for a diagnosis of RA [11] and were recruited between March 2009 and September 2011. All cases had active disease at the time of blood sampling: Disease Activity Score (DAS) 28 of 5.55 (range of 4.07 to 8.26). The patients maintained the same doses of methotrexate no less than 3 months and did not concurrently receive other disease-modifying anti-rheumatic drugs or biologics before the study. The baseline demographic characteristics of patients and healthy controls are detailed in Table S1 (Additional file 1). The study was approved by the Medical Ethics Committee of Peking University People’s Hospital, and informed consent was obtained from all participants.

CD4+ T-cell RNA processing

In total, 12-mL whole blood samples were drawn and stored at 4°C for less than 4 hours. Samples were layered onto Ficoll-Paque Plus, and PBMCs were separated by density gradient centrifugation. An automated magnetic beadbased-positive selection protocol was used to isolate CD4 cells (Stemcell Technologies, Vancouver, BC, Canada). By this approach, a median purity of 95% CD4+ T cells was achieved by flow cytometry analysis. Total CD4+ T-cell RNA was extracted by using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) and purified by using an RNeasy Mini Kit (Qiagen, Hilden, Germany). Only RNA preparations with a 28S/18S ratio of more than 1.7 and an A260/280 range of 1.8 to approximately 2.1 were used for gene expression analysis.

Microarray assay

The transcriptome analyses used Human Genome U133 Plus version 2.0 high-density oligonucleotide arrays (Affymetrix, Santa Clara, CA, USA) with 54,000 probe sets and 1,300,000 distinct oligonucleotides to interrogate 47,000 well-characterized human transcripts. The sample labeling, microarray hybridization, and washing were performed on the basis of the standard protocols of the manufacturer. Briefly, total RNA was transcribed to double-strand cDNA and then to synthesized cRNA and labeled with cyanine-3-CTP. The labeled cRNAs were hybridized onto the microarray. After the slides were washed, the arrays were scanned by GeneChip Scanner 3000 (Affymetrix). The gene expression array data were digitalized by using GeneChip Operating Software (version 1.4; Affymetrix) and normalized by eliminating the highest and lowest 2% of the data by using MAS5 algorithm (Affymetrix). The microarray data have been deposited in the Gene Expression Omnibus (GEO) of the National Center for Biotechnology Information (NCBI) and are accessible through GEO series accession number GSE56649.

Real-time quantitative polymerase chain reaction

The selected candidate genes were validated by qPCR. Briefly, the cDNA was synthesized in accordance with the instructions indicated in a RevertAid™ First-Strand cDNA Synthesis Kit (Fermentas, Shenzhen, China). Two-step PCR was performed by using SYBR Green PCR Master Mix (Applied Biosystems, Foster City, CA, USA) in accordance with the instructions of the manufacturer. The reaction was run on an ABI 9700 fluorescent sequence detection system (Applied Biosystems). Gene expression was quantified relative to the expression of the housekeeping gene 18 s rRNA and normalized to control by standard 2−△△CT calculation. Primer sequences used are summarized in Table S2 (Additional file 2).

Statistical analysis

For microarray analysis, a differentially expressed (DE) gene was defined if its geometric mean of intensities reached at least 1.5-fold changes between case and control groups. The false-discovery rate (FDR) was applied to determine the statistical significance. An FDR-adjusted P value (q value) of below 0.05 was defined as statistically significant. The independent-samples t test was applied for the analysis of candidate gene validation, and a P value of less than 0.05 was considered statistically significant after Bonferroni correction. All statistical analyses were conducted by using program SPSS 13.0 (SPSS Inc., Chicago, IL, USA).

Gene Ontology, gene-gene network, and pathway analysis

The significant DE genes from microarray were first analyzed for hierarchical clustering (Cluster 3.0, available at [12]) and visualized with Treeview 3.0 (available at [13]). To interpret biological meaning of the transcripts, the DE genes were functionally categorized according to the Gene Ontology (GO) database [14]. GO analysis allowed us to classify the large gene list into functionally related gene sets according to a reference (NCBI: Homo sapiens genes). Fisher’s exact test and chi-squared test were used to statistically classify GO categories, and an FDR (q value) of less than 0.05 was considered statistically significant. The gene-gene interaction networks and pathway analysis were analyzed by using Ingenuity Pathway Analysis software version 7.5 [15]. Interactions among the DE genes were investigated according to the Human Protein Reference Database (HPRD) [16] and the Molecular INTeraction (MINT) database [17]. The significance of the interactions was presented as gene modules and evaluated according to between-ness centrality reflecting the importance of a module in relation to other modules [18-20]. Pathway analysis was applied to find out the significant pathways among DE genes according to the Kyoto Encyclopedia of Genes and Genomes (KEGG) [21]. Pathways generated by fewer than five uploaded genes were excluded from the analysis. Pathway enrichment analysis (PEA) (those with the highest percentage of genes in a particular pathway) was applied to evaluate the significance of pathways [22-24].

Results

CD4 T-cell transcriptome in patients with active rheumatoid arthritis versus healthy controls

The transcriptome profiles of CD4 T cells from 13 cases with active RA and nine healthy controls were accessed by microarrays. Using a q value of less than 0.05 and taking into account genes with at least 1.5-fold changes, we identified a total of 1,496 DE transcripts. The proportion of DE transcripts was substantially higher among RefSeq genes (89%) than non-RefSeq genes (11%). Among the transcripts, 832 were upregulated (Table S3 in Additional file 3) and the remaining 664 were downregulated (Table S4 in Additional file 4). These genes clearly separated patients with active RA from healthy individuals and were visualized in a hierarchical clustering diagram (Figure 1a).

Gene Ontology analysis of differentially expressed transcripts defined five major gene categories

By GO analysis, the common genes can be generally classified into three GO systems: biological process, cellular component and molecular function, and (more specifically) ‘higher level’ descriptive GO terms (for example, ‘signal transduction’ or ‘regulation of apoptosis’). We found that the DE genes in CD4 T cells were enriched in several common ontologies, including immune response (GO:0006955, 122 genes, q = 5.99 × 10−8), more specifically in (i) regulation of adaptive immune response (GO:0002819); (ii) positive regulation of chronic inflammatory response (GO:0002678); (iii) interferon-gamma (IFNγ)-mediated signaling pathway (GO:0060333); and (iv) response to cytokine stimulus (GO:0034097), apoptosis process (111 genes, q = 1.73 × 10−5), more specifically in (i) induction of apoptosis by intracellular signals (GO:0008629); (ii) cellular component involved in apoptosis (GO:0006921); (iii) regulation of myeloid cell apoptosis (GO:0033032); and (iv) recognition of apoptotic cell (GO:0043654), T-cell receptor (TCR) signaling pathway (GO:0050852, 40 genes, q = 2.98 × 10−6), and regulation of kinase (GO:0007243, 58 genes, q = 3.29 × 10−4) and phosphatase (GO:0016310, 31 genes, q = 9.36 × 10−4) activity (Figure 1b-e, Table S5-8 in Additional files 5, 6, 7 and 8). Interestingly, of the significantly enriched GO categories, one was related to the Wnt receptor signaling pathway (GO:0016055, 23 genes, q = 4.15 × 10−4) (Figure 1f, Table S9 in Additional file 9). A similar result was obtained by pathway analysis, which identified ‘Wnt signaling pathway’ with great significance of differential regulation between two groups (q = 2.78 × 10−10). The five major GO terms are summarized in Table 1.
Table 1
Gene Ontology in differentially expressed genes
GO_Id
GO_term
Count
Q value
GO:0006955
Immune response
122
5.99 × 10−8
GO:0002819
Regulation of adaptive immune response
23
9.37 × 10−4
GO:0002678
Positive regulation of chronic inflammatory response
12
1.38 × 10−3
GO:0060333
Interferon-gamma-mediated signaling pathway
9
6.02 × 10−3
GO:0034097
Response to cytokine stimulus
17
8.61 × 10−3
GO:0006915
Apoptosis
111
1.73 × 10−5
GO:0008629
Induction of apoptosis by intracellular signals
17
4.15 × 10−4
GO:0006921
Cellular component involved in apoptosis
29
2.38 × 10−3
GO:0033032
Regulation of myeloid cell apoptosis
22
7.18 × 10−3
GO:0043654
Recognition of apoptotic cell
7
8.42 × 10−2
GO:0050852
T-cell receptor signaling pathway
40
2.98 × 10−6
GO:0007243
Intracellular protein kinase cascade
58
3.29 × 10−4
GO:0016310
Phosphorylation
31
9.36 × 10−4
GO:0016055
Wnt receptor signaling pathway
23
4.15 × 10−4

Corresponding interaction networks among differentially expressed genes in the five major Gene Ontology categories

To identify more significant DE genes, the network interactions among DE genes implicated in the five major GO categories were investigated according to HPRD and MINT databases. As shown in Table S10 (Additional file 10), the DE gene modules, such as PIK3R1, PAK1, CBL, PIK3CA, SYK, and ZAP70, were highly relevant to TCR signaling and JAK-STAT signaling. A cluster of genes involved in Wnt signaling (for example, TLE1, CSNK2A1, and MAPK8) were also present in this network. Additionally, several gene modules with possible functional involvement in RA pathogenesis, such as ZAP70, HSP90AA1, HSP90AB1, and VIM, were depicted in the network.

Corresponding signaling pathways of differentially expressed genes

The possible signaling pathways implicated in regulation of CD4 T cells in active RA were investigated according to the KEGG database. The high-ranked pathways are displayed in Table 2. The DE genes were highly relevant to T-cell response, such as the ErbB signaling pathway (PEA = 28.122, q = 4.88 × 10−14), the TCR signaling pathway (PEA = 24.542, q = 2.69 × 10−14), the mammalian target of rapamycin (mTOR) signaling pathway (PEA = 23.525, q = 1.71 × 10−7), and the TGF-β signaling pathway (PEA = 23.435, q = 2.63 × 10−11). In addition, several canonical pathways were depicted, such as apoptosis (PEA = 22.909, q = 3.01 × 10−11), the chemokine signaling pathway (PEA = 19.114, q = 1.52 × 10−17), the MAPK signaling pathway (PEA = 17.304, P = 2.14 × 10−21), and the Jak-STAT signaling pathway (PEA = 15.785, q = 2.44 × 10−11). Interestingly, the Wnt signaling pathway (PEA = 14.755, q = 2.78 × 10−10) and the systemic lupus erythematosus (SLE) signaling pathway (PEA = 7.282, q = 3.47 × 10−4) were also depicted in this analysis. Furthermore, by pathway network analysis, several genes, such as MAPK1, PIK3CA, PIK3R1, KRAS, and PRKCB, were centered in the network (Figure 2). The majority of these genes overlapped with the ones in the interaction network analysis (Table S11, Additional file 11).
Table 2
Identification of signaling pathways based on Kyoto Encyclopedia of Genes and Genomes in differentially expressed genes
Path_Id
Path_name
Path_diffgenes
Path_genes
Enrichment
Q value
4012
ErbB signaling pathway
12
87
28.122
4.88 × 10−14
4660
T-cell receptor signaling pathway
13
108
24.542
2.69 × 10−14
4150
mTOR signaling pathway
6
52
23.525
1.71 × 10−7
4350
TGF-β signaling pathway
10
87
23.435
2.63 × 10−11
4210
Apoptosis
10
89
22.909
3.01 × 10−11
4062
Chemokine signaling pathway
18
192
19.114
1.52 × 10−17
4010
MAPK signaling pathway
23
271
17.304
2.14 × 10−21
4630
Jak-STAT signaling pathway
12
155
15.785
2.44 × 10−11
4310
Wnt signaling pathway
11
152
14.755
2.78 × 10−10
4620
Toll-like receptor signaling pathway
5
101
10.093
9.23 × 10−5
4060
Cytokine-cytokine receptor interaction
12
263
9.303
6.59 × 10−9
5322
Systemic lupus erythematosus
5
140
7.282
3.47 × 10−4
Finally, we performed gene module analysis to combine the results from GO, gene-gene interaction, and pathway analysis and found that a substantial number of genes in the three different analyses overlapped. In total, 23 genes were selected for further validation using a cut-off q value of 0.05 and an exclusion of being extensively studied in RA according to publications. These genes are characterized in Table 3.
Table 3
Gene modules resulting from the combination of Gene Ontology, gene-gene interaction, and pathway analysis
Probe set ID
Gene symbol
Fold change
Q value
229010_at
CBL
2.22
0
212435_at
TRIM33
0.61
0
226294_x_at
FAM91A1
0.33
0
227817_at
PRKCB
2.26
6.101 × 10−16
227073_at
MAP3K2
0.50
1.296 × 10−14
214578_s_at
ROCK1
2.99
4.303 × 10−12
212073_at
CSNK2A1
0.32
5.813 × 10−8
1555613_a_at
ZAP70
0.59
6.82 × 10−6
223300_s_at
CCDC82
0.51
6.82 × 10−6
212366_at
ZNF292
0.59
2.31 × 10−5
214917_at
PRKAA1
0.57
7.15 × 10−5
212249_at
PIK3R1
0.58
0.000141
231777_at
CSNK2B
3.90
0.00076
37152_at
PPARD
0.44
0.000927
231798_at
NOG
1.62
0.00133
225661_at
IFNAR1
2.16
0.002
235980_at
PIK3CA
1.70
0.00264
206359_at
SOCS3
0.62
0.00336
226660_at
RPS6KB1
0.61
0.0101
212764_at
ZEB1
1.83
0.0125
1553725_s_at
ZNF644
0.59
0.0187
203542_s_at
KLF9
0.61
0.0238
211930_at
HNRNPA3
0.64
0.0344

Validation analysis confirmed that STAT3 profiling and Wnt signaling are the prominent active rheumatoid arthritis ‘signature’

qPCR was used to validate the selected 23 candidate genes. As shown in Figure 3, a robust differential expression was confirmed for eight of the 23 genes. One gene, PIK3CA, exhibited similar expression patterns but did not reach the statistical significance (P = 0.055). Interestingly, four out of the nine genes (SOCS3, CBL, IFNAR1, and PIK3CA) were noted to have STAT3 signaling involvement, and three genes (CBL, KLF9, and CSNK2A1) were involved in Wnt signaling pathway, based on the KEGG database and recent publications [25,26]. Additionally, several zinc finger transcription factors (ZEB1, ZNF292, and ZNF644) were confirmed.

Discussion

RA is a chronic autoimmune disorder in which T cells play a pivotal role in the initiation and progression of the disease. Of these, CD4 T cells are key players in RA pathogenesis. A previous microarray study was conducted on CD4 T cells during the early phase of RA in a case-only design [10]. However, to date, no studies have focused on the expression profile of CD4 T cells between patients with RA and healthy individuals. Herein, we performed a unique CD4 T-cell transcriptome analysis in a case–control cohort. For the first time, we showed that CD4 T cells from patients with active RA and those from healthy controls had distinct gene regulations.
Our data indicate that the DE genes between two groups mainly involved in regulation of immune responses, especially T-cell response, and other cellular processes such as kinase and phosphatase regulator activity. There were also a significant number of genes involved in regulation of apoptosis. Apoptosis is a key process regulating immune homeostasis. Abnormalities in T-cell apoptosis resulted in a wide range of pathologic conditions. For instance, infection of CD4 T cells with HIV results in depletion of these lymphocytes because of programmed cell death [27]. In RA, one of the principal characteristics is ‘apoptosis resistance’ in the synovial tissues [28]. The synovial fluid T cells from the patients with RA were resistant to FasL-induced apoptosis [29]. Furthermore, accumulated evidence suggested that the Wnt signaling pathway plays an important role in FLS activation, bone resorption, and joint destruction in RA pathogenesis [30-34]. In the context of Wnt signaling in T-cell activation, it has been reported that activation of Wnt signaling in CD4+CD25+ regulatory T (Treg) cells increased the survival of Treg cells and induced an anergic phenotype in CD4+CD25 effector T cells [35,36]. However, so far, few studies have focused on the involvement of Wnt signaling in T cells in RA development. Lee et al. [37] reported that sFRP1 (secreted frizzled-related protein 1), an inhibitor of Wnt signaling, was correlated with interleukin-17 (IL-17) levels in RA synovial fluid. In vitro, overexpression of sFRP1 induced a significant increase of T helper 17 (Th17) cells. In the present study, we observed that one of the significant GO categories was Wnt receptor signaling. Pathway analysis also indicates that Wnt signaling was one of the most significant pathways in the regulation of RA activity. Our findings support the notion that the Wnt signaling pathway may participate in the impaired T-cell homeostasis in RA. Notably, the genes involved in the SLE signaling pathway were also enriched in the present study, indicating that some of the genes are commonly shared by multiple autoimmune diseases, such as RA and SLE.
To reduce the complexity of the whole-genome expression data and to determine the most significant DE genes, we performed the gene module analysis to combine the results from GO, gene-gene interaction, and pathway analysis. In total, 23 genes were selected for further validation using a cut-off q value of 0.05 and an exclusion of being extensively studied in RA according to publications. Nine of those genes were confirmed in the validation cohort. Given the prominent importance of the STAT3 signaling pathway in many cellular functions, including T-cell differentiation [38], it is notable that four of the nine DE genes (SOCS3, CBL, IFNAR1, and PIK3CA) were involved in STAT3 signaling. SOCS3 is a key negative regulator that inhibits the STAT3 signaling pathway and is a major negative regulator of CD4 T-cell activation [39]. Huang et al. [40] reported that the expression of SOCS3 was elevated in patients with hepatitis C virus. The activation of SOCS3 contributes to the defective hepatic response to IFNγ. By contrast, reduced expression of SOCS3 has been observed in various human inflammatory diseases. Mice lacking Socs3 developed an exacerbated inflammatory arthritis and were characterized by increased numbers of Th17 cells [41]. In the present work, SOCS3 expression was downregulated in RA CD4 T cells. The finding was consistent with a previous observation of the involvement of STAT3 signaling in patients with early arthritis [10]. Another interesting observation is that three validated transcripts (CBL, KLF9, and CSNK2A1) were dominantly related to Wnt signaling pathways. CBL is originally identified as a RING finger ubiquitin E3 ligase and recently has emerged as a key regulator of Wnt signaling by targeting the active β-catenin [25]. KLF9 is a transcriptional regulator of cell proliferation, adhesion, and differentiation and recently was shown to be a negative regulator of putative Wnt inhibitor DKK1 promoter activity in human stromal cells [26]. Furthermore, KLF9 was shown to function as a suppressor of tumor-initiating stem cells by directly suppressing Notch1 signaling [42]. As compelling evidence supports Notch involvement in CD4 T-cell differentiation [43-46], we speculate that KLF9 may function as a suppressor of CD4 T cells via the Notch signaling pathway and play a role in RA pathogenesis. Further functional study of the DE gene is needed to fully understand its contribution to RA. In addition, three zinc finger transcription factors (ZEB1, ZNF292, and ZNF644) were confirmed. Zinc finger proteins are the most abundant proteins in eukaryotes and play an important role in various cellular processes. Their functions are extremely diverse, including DNA recognition, apoptosis regulation, and transcriptional activation. Wang et al. [47] reported that ZEB1 acts as a specific repressor of IL-2 gene transcription and functions. Overexpression of ZEB1 can repress IL-2 promoter activity and endogenous IL-2 production in T cells. However, little is known about the role of the three zinc finger transcription factors in autoimmune diseases. Our results warrant the functional characterization of these zinc finger molecules to fully understand their contribution to RA pathogenesis.

Conclusions

We report here the first case–control study of CD4 T-cell transcriptome profile in RA. Our data provide evidence that CD4 T cells from patients with RA had abnormal functional networks in the STAT3 signaling pathway and Wnt signaling. Our results also suggest that the aberrant expression of several zinc finger transcription factors (ZEB1, ZNF292, and ZNF644) may be potential pathogenic factors for RA.

Acknowledgments

We thank the staff of the Department of Rheumatology and Immunology, People’s Hospital, Peking University, for recruiting patients and healthy controls and for the technical assistance. We thank the patients and healthy volunteers for their cooperation and for consenting to participate in this study. This study is supported partly by the National Basic Research Program of China (Program 973) (#2010CB529105), the National Natural Science Foundation of China (#81120108020, #31170840, and #31270914), the Beijing Natural Science Foundation (#7122192 and #7122196), Peking University ‘Project 985’, and the Scientific Research Foundation for the Returned Overseas Chinese Scholars, State Education Ministry (#JWSL431).

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HY contributed to the study design, clinical data collection, data interpretation, and manuscript drafting. JZ carried out real-time polymerase chain reaction experiments, participated in statistical analysis, and helped to revise the manuscript. JW helped to perform the bioinformatic analysis and revised the manuscript. YG, YD, and CL helped in the samples collection, clinical data interpretation, and manuscript revision. MD helped in the bioinformatic and statistical analysis and revised the manuscript. JG contributed to the study design and carried out the bioinformatic and statistical analysis, data interpretation, and manuscript drafting and revision. ZL conceived of the study, participated in its design, and helped to revise the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Kallberg H, Padyukov L, Plenge RM, Ronnelid J, Gregersen PK, van der Helm-van Mil AH, et al. Gene-gene and gene-environment interactions involving HLA-DRB1, PTPN22, and smoking in two subsets of rheumatoid arthritis. Am J Hum Genet. 2007;80:867–75.CrossRefPubMedCentralPubMed Kallberg H, Padyukov L, Plenge RM, Ronnelid J, Gregersen PK, van der Helm-van Mil AH, et al. Gene-gene and gene-environment interactions involving HLA-DRB1, PTPN22, and smoking in two subsets of rheumatoid arthritis. Am J Hum Genet. 2007;80:867–75.CrossRefPubMedCentralPubMed
3.
Zurück zum Zitat Burska AN, Roget K, Blits M, Soto Gomez L, van de Loo F, Hazelwood LD, et al. Gene expression analysis in RA: towards personalized medicine. Pharmacogenomics J. 2014;14:93–106.CrossRefPubMedCentralPubMed Burska AN, Roget K, Blits M, Soto Gomez L, van de Loo F, Hazelwood LD, et al. Gene expression analysis in RA: towards personalized medicine. Pharmacogenomics J. 2014;14:93–106.CrossRefPubMedCentralPubMed
4.
Zurück zum Zitat Tsubaki T, Arita N, Kawakami T, Shiratsuchi T, Yamamoto H, Takubo N, et al. Characterization of histopathology and gene-expression profiles of synovitis in early rheumatoid arthritis using targeted biopsy specimens. Arthritis Res Ther. 2005;7:R825–36.CrossRefPubMedCentralPubMed Tsubaki T, Arita N, Kawakami T, Shiratsuchi T, Yamamoto H, Takubo N, et al. Characterization of histopathology and gene-expression profiles of synovitis in early rheumatoid arthritis using targeted biopsy specimens. Arthritis Res Ther. 2005;7:R825–36.CrossRefPubMedCentralPubMed
5.
Zurück zum Zitat Galligan CL, Baig E, Bykerk V, Keystone EC, Fish EN. Distinctive gene expression signatures in rheumatoid arthritis synovial tissue fibroblast cells: correlates with disease activity. Genes Immun. 2007;8:480–91.CrossRefPubMed Galligan CL, Baig E, Bykerk V, Keystone EC, Fish EN. Distinctive gene expression signatures in rheumatoid arthritis synovial tissue fibroblast cells: correlates with disease activity. Genes Immun. 2007;8:480–91.CrossRefPubMed
6.
Zurück zum Zitat Badot V, Galant C, Nzeusseu Toukap A, Theate I, Maudoux AL, Van den Eynde BJ, et al. Gene expression profiling in the synovium identifies a predictive signature of absence of response to adalimumab therapy in rheumatoid arthritis. Arthritis Res Ther. 2009;11:R57.CrossRefPubMedCentralPubMed Badot V, Galant C, Nzeusseu Toukap A, Theate I, Maudoux AL, Van den Eynde BJ, et al. Gene expression profiling in the synovium identifies a predictive signature of absence of response to adalimumab therapy in rheumatoid arthritis. Arthritis Res Ther. 2009;11:R57.CrossRefPubMedCentralPubMed
7.
Zurück zum Zitat Teixeira VH, Olaso R, Martin-Magniette ML, Lasbleiz S, Jacq L, Oliveira CR, et al. Transcriptome analysis describing new immunity and defense genes in peripheral blood mononuclear cells of rheumatoid arthritis patients. PLoS One. 2009;4:e6803.CrossRefPubMedCentralPubMed Teixeira VH, Olaso R, Martin-Magniette ML, Lasbleiz S, Jacq L, Oliveira CR, et al. Transcriptome analysis describing new immunity and defense genes in peripheral blood mononuclear cells of rheumatoid arthritis patients. PLoS One. 2009;4:e6803.CrossRefPubMedCentralPubMed
8.
Zurück zum Zitat Lequerre T, Bansard C, Vittecoq O, Derambure C, Hiron M, Daveau M, et al. Early and long-standing rheumatoid arthritis: distinct molecular signatures identified by gene-expression profiling in synovia. Arthritis Res Ther. 2009;11:R99.CrossRefPubMedCentralPubMed Lequerre T, Bansard C, Vittecoq O, Derambure C, Hiron M, Daveau M, et al. Early and long-standing rheumatoid arthritis: distinct molecular signatures identified by gene-expression profiling in synovia. Arthritis Res Ther. 2009;11:R99.CrossRefPubMedCentralPubMed
9.
Zurück zum Zitat Lee HM, Sugino H, Aoki C, Shimaoka Y, Suzuki R, Ochi K, et al. Abnormal networks of immune response-related molecules in bone marrow cells from patients with rheumatoid arthritis as revealed by DNA microarray analysis. Arthritis Res Ther. 2011;13:R89.CrossRefPubMedCentralPubMed Lee HM, Sugino H, Aoki C, Shimaoka Y, Suzuki R, Ochi K, et al. Abnormal networks of immune response-related molecules in bone marrow cells from patients with rheumatoid arthritis as revealed by DNA microarray analysis. Arthritis Res Ther. 2011;13:R89.CrossRefPubMedCentralPubMed
10.
Zurück zum Zitat Pratt AG, Swan DC, Richardson S, Wilson G, Hilkens CM, Young DA, et al. A CD4 T cell gene signature for early rheumatoid arthritis implicates interleukin 6-mediated STAT3 signalling, particularly in anti-citrullinated peptide antibody-negative disease. Ann Rheum Dis. 2012;71:1374–81.CrossRefPubMedCentralPubMed Pratt AG, Swan DC, Richardson S, Wilson G, Hilkens CM, Young DA, et al. A CD4 T cell gene signature for early rheumatoid arthritis implicates interleukin 6-mediated STAT3 signalling, particularly in anti-citrullinated peptide antibody-negative disease. Ann Rheum Dis. 2012;71:1374–81.CrossRefPubMedCentralPubMed
11.
Zurück zum Zitat Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31:315–24.CrossRefPubMed Arnett FC, Edworthy SM, Bloch DA, McShane DJ, Fries JF, Cooper NS, et al. The American Rheumatism Association 1987 revised criteria for the classification of rheumatoid arthritis. Arthritis Rheum. 1988;31:315–24.CrossRefPubMed
17.
Zurück zum Zitat Licata L, Briganti L, Peluso D, Perfetto L, Iannuccelli M, Galeota E, et al. MINT, the molecular interaction database: 2012 update. Nucleic Acids Res. 2012;40(Database issue):D857–61. Licata L, Briganti L, Peluso D, Perfetto L, Iannuccelli M, Galeota E, et al. MINT, the molecular interaction database: 2012 update. Nucleic Acids Res. 2012;40(Database issue):D857–61.
18.
20.
Zurück zum Zitat Li C, Li H. Network-constrained regularization and variable selection for analysis of genomic data. Bioinformatics. 2008;24:1175–82.CrossRefPubMed Li C, Li H. Network-constrained regularization and variable selection for analysis of genomic data. Bioinformatics. 2008;24:1175–82.CrossRefPubMed
22.
23.
Zurück zum Zitat Yi M, Horton JD, Cohen JC, Hobbs HH, Stephens RM. WholePathwayScope: a comprehensive pathway-based analysis tool for high-throughput data. BMC Bioinformatics. 2006;7:30.CrossRefPubMedCentralPubMed Yi M, Horton JD, Cohen JC, Hobbs HH, Stephens RM. WholePathwayScope: a comprehensive pathway-based analysis tool for high-throughput data. BMC Bioinformatics. 2006;7:30.CrossRefPubMedCentralPubMed
24.
Zurück zum Zitat Draghici S, Khatri P, Tarca AL, Amin K, Done A, Voichita C, et al. A systems biology approach for pathway level analysis. Genome Res. 2007;17:1537–45.CrossRefPubMedCentralPubMed Draghici S, Khatri P, Tarca AL, Amin K, Done A, Voichita C, et al. A systems biology approach for pathway level analysis. Genome Res. 2007;17:1537–45.CrossRefPubMedCentralPubMed
25.
Zurück zum Zitat Chitalia V, Shivanna S, Martorell J, Meyer R, Edelman E, Rahimi N. c-Cbl, a ubiquitin E3 ligase that targets active beta-catenin: a novel layer of Wnt signaling regulation. J Biol Chem. 2013;288:23505–17.CrossRefPubMed Chitalia V, Shivanna S, Martorell J, Meyer R, Edelman E, Rahimi N. c-Cbl, a ubiquitin E3 ligase that targets active beta-catenin: a novel layer of Wnt signaling regulation. J Biol Chem. 2013;288:23505–17.CrossRefPubMed
26.
Zurück zum Zitat Pabona JM, Simmen FA, Nikiforov MA, Zhuang D, Shankar K, Velarde MC, et al. Kruppel-like factor 9 and progesterone receptor coregulation of decidualizing endometrial stromal cells: implications for the pathogenesis of endometriosis. J Clin Endocrinol Metab. 2012;97:E376–92.CrossRefPubMedCentralPubMed Pabona JM, Simmen FA, Nikiforov MA, Zhuang D, Shankar K, Velarde MC, et al. Kruppel-like factor 9 and progesterone receptor coregulation of decidualizing endometrial stromal cells: implications for the pathogenesis of endometriosis. J Clin Endocrinol Metab. 2012;97:E376–92.CrossRefPubMedCentralPubMed
27.
Zurück zum Zitat Schattner E, Laurence J. HIV-induced T-lymphocyte depletion. Clin Lab Med. 1994;14:221–38.PubMed Schattner E, Laurence J. HIV-induced T-lymphocyte depletion. Clin Lab Med. 1994;14:221–38.PubMed
28.
Zurück zum Zitat Liu H, Pope RM. Apoptosis in rheumatoid arthritis: friend or foe. Rheum Dis Clin North Am. 2004;30:603–25. x. Liu H, Pope RM. Apoptosis in rheumatoid arthritis: friend or foe. Rheum Dis Clin North Am. 2004;30:603–25. x.
29.
Zurück zum Zitat Martinez-Lorenzo MJ, Anel A, Saez-Gutierrez B, Royo-Canas M, Bosque A, Alava MA, et al. Rheumatoid synovial fluid T cells are sensitive to APO2L/TRAIL. Clin Immunol. 2007;122:28–40.CrossRefPubMed Martinez-Lorenzo MJ, Anel A, Saez-Gutierrez B, Royo-Canas M, Bosque A, Alava MA, et al. Rheumatoid synovial fluid T cells are sensitive to APO2L/TRAIL. Clin Immunol. 2007;122:28–40.CrossRefPubMed
30.
Zurück zum Zitat Sonomoto K, Yamaoka K, Oshita K, Fukuyo S, Zhang X, Nakano K, et al. Interleukin-1beta induces differentiation of human mesenchymal stem cells into osteoblasts via the Wnt-5a/receptor tyrosine kinase-like orphan receptor 2 pathway. Arthritis Rheum. 2012;64:3355–63.CrossRefPubMed Sonomoto K, Yamaoka K, Oshita K, Fukuyo S, Zhang X, Nakano K, et al. Interleukin-1beta induces differentiation of human mesenchymal stem cells into osteoblasts via the Wnt-5a/receptor tyrosine kinase-like orphan receptor 2 pathway. Arthritis Rheum. 2012;64:3355–63.CrossRefPubMed
31.
Zurück zum Zitat Chan BY, Little CB. The interaction of canonical bone morphogenetic protein- and Wnt-signaling pathways may play an important role in regulating cartilage degradation in osteoarthritis. Arthritis Res Ther. 2012;14:119.CrossRefPubMedCentralPubMed Chan BY, Little CB. The interaction of canonical bone morphogenetic protein- and Wnt-signaling pathways may play an important role in regulating cartilage degradation in osteoarthritis. Arthritis Res Ther. 2012;14:119.CrossRefPubMedCentralPubMed
32.
Zurück zum Zitat Maeda K, Kobayashi Y, Udagawa N, Uehara S, Ishihara A, Mizoguchi T, et al. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nat Med. 2012;18:405–12.CrossRefPubMed Maeda K, Kobayashi Y, Udagawa N, Uehara S, Ishihara A, Mizoguchi T, et al. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nat Med. 2012;18:405–12.CrossRefPubMed
33.
Zurück zum Zitat de Rooy DP, Yeremenko NG, Wilson AG, Knevel R, Lindqvist E, Saxne T, et al. Genetic studies on components of the Wnt signalling pathway and the severity of joint destruction in rheumatoid arthritis. Ann Rheum Dis. 2013;72:769–75.CrossRefPubMed de Rooy DP, Yeremenko NG, Wilson AG, Knevel R, Lindqvist E, Saxne T, et al. Genetic studies on components of the Wnt signalling pathway and the severity of joint destruction in rheumatoid arthritis. Ann Rheum Dis. 2013;72:769–75.CrossRefPubMed
34.
Zurück zum Zitat Shin Y, Huh YH, Kim K, Kim S, Park KH, Koh JT, et al. Low-density lipoprotein receptor-related protein 5 governs Wnt-mediated osteoarthritic cartilage destruction. Arthritis Res Ther. 2014;16:R37.CrossRefPubMedCentralPubMed Shin Y, Huh YH, Kim K, Kim S, Park KH, Koh JT, et al. Low-density lipoprotein receptor-related protein 5 governs Wnt-mediated osteoarthritic cartilage destruction. Arthritis Res Ther. 2014;16:R37.CrossRefPubMedCentralPubMed
35.
Zurück zum Zitat Staal FJ, Luis TC, Tiemessen MM. WNT signalling in the immune system: WNT is spreading its wings. Nat Rev Immunol. 2008;8:581–93.CrossRefPubMed Staal FJ, Luis TC, Tiemessen MM. WNT signalling in the immune system: WNT is spreading its wings. Nat Rev Immunol. 2008;8:581–93.CrossRefPubMed
36.
Zurück zum Zitat Ding Y, Shen S, Lino AC. Curotto de Lafaille MA, Lafaille JJ: Beta-catenin stabilization extends regulatory T cell survival and induces anergy in nonregulatory T cells. Nat Med. 2008;14:162–9.CrossRefPubMed Ding Y, Shen S, Lino AC. Curotto de Lafaille MA, Lafaille JJ: Beta-catenin stabilization extends regulatory T cell survival and induces anergy in nonregulatory T cells. Nat Med. 2008;14:162–9.CrossRefPubMed
37.
Zurück zum Zitat Lee YS, Lee KA, Yoon HB, Yoo SA, Park YW, Chung Y, et al. The Wnt inhibitor secreted Frizzled-Related Protein 1 (sFRP1) promotes human Th17 differentiation. Eur J Immunol. 2012;42:2564–73.CrossRefPubMed Lee YS, Lee KA, Yoon HB, Yoo SA, Park YW, Chung Y, et al. The Wnt inhibitor secreted Frizzled-Related Protein 1 (sFRP1) promotes human Th17 differentiation. Eur J Immunol. 2012;42:2564–73.CrossRefPubMed
39.
Zurück zum Zitat Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7:454–65.CrossRefPubMed Yoshimura A, Naka T, Kubo M. SOCS proteins, cytokine signalling and immune regulation. Nat Rev Immunol. 2007;7:454–65.CrossRefPubMed
40.
Zurück zum Zitat Huang Y, Feld JJ, Sapp RK, Nanda S, Lin JH, Blatt LM, et al. Defective hepatic response to interferon and activation of suppressor of cytokine signaling 3 in chronic hepatitis C. Gastroenterology. 2007;132:733–44.CrossRefPubMedCentralPubMed Huang Y, Feld JJ, Sapp RK, Nanda S, Lin JH, Blatt LM, et al. Defective hepatic response to interferon and activation of suppressor of cytokine signaling 3 in chronic hepatitis C. Gastroenterology. 2007;132:733–44.CrossRefPubMedCentralPubMed
41.
Zurück zum Zitat Wong PK, Egan PJ, Croker BA, O’Donnell K, Sims NA, Drake S, et al. SOCS-3 negatively regulates innate and adaptive immune mechanisms in acute IL-1-dependent inflammatory arthritis. J Clin Invest. 2006;116:1571–81.CrossRefPubMedCentralPubMed Wong PK, Egan PJ, Croker BA, O’Donnell K, Sims NA, Drake S, et al. SOCS-3 negatively regulates innate and adaptive immune mechanisms in acute IL-1-dependent inflammatory arthritis. J Clin Invest. 2006;116:1571–81.CrossRefPubMedCentralPubMed
42.
Zurück zum Zitat Ying M, Sang Y, Li Y, Guerrero-Cazares H, Quinones-Hinojosa A, Vescovi AL, et al. Kruppel-like family of transcription factor 9, a differentiation-associated transcription factor, suppresses Notch1 signaling and inhibits glioblastoma-initiating stem cells. Stem Cells. 2011;29:20–31.CrossRefPubMedCentralPubMed Ying M, Sang Y, Li Y, Guerrero-Cazares H, Quinones-Hinojosa A, Vescovi AL, et al. Kruppel-like family of transcription factor 9, a differentiation-associated transcription factor, suppresses Notch1 signaling and inhibits glioblastoma-initiating stem cells. Stem Cells. 2011;29:20–31.CrossRefPubMedCentralPubMed
43.
Zurück zum Zitat Bailis W, Yashiro-Ohtani Y, Fang TC, Hatton RD, Weaver CT, Artis D, et al. Notch simultaneously orchestrates multiple helper T cell programs independently of cytokine signals. Immunity. 2013;39:148–59.CrossRefPubMedCentralPubMed Bailis W, Yashiro-Ohtani Y, Fang TC, Hatton RD, Weaver CT, Artis D, et al. Notch simultaneously orchestrates multiple helper T cell programs independently of cytokine signals. Immunity. 2013;39:148–59.CrossRefPubMedCentralPubMed
44.
Zurück zum Zitat Keerthivasan S, Suleiman R, Lawlor R, Roderick J, Bates T, Minter L, et al. Notch signaling regulates mouse and human Th17 differentiation. J Immunol. 2011;187:692–701.CrossRefPubMedCentralPubMed Keerthivasan S, Suleiman R, Lawlor R, Roderick J, Bates T, Minter L, et al. Notch signaling regulates mouse and human Th17 differentiation. J Immunol. 2011;187:692–701.CrossRefPubMedCentralPubMed
45.
Zurück zum Zitat Amsen D, Antov A, Flavell RA. The different faces of Notch in T-helper-cell differentiation. Nat Rev Immunol. 2009;9:116–24.CrossRefPubMed Amsen D, Antov A, Flavell RA. The different faces of Notch in T-helper-cell differentiation. Nat Rev Immunol. 2009;9:116–24.CrossRefPubMed
46.
Zurück zum Zitat Amsen D, Antov A, Jankovic D, Sher A, Radtke F, Souabni A, et al. Direct regulation of Gata3 expression determines the T helper differentiation potential of Notch. Immunity. 2007;27:89–99.CrossRefPubMedCentralPubMed Amsen D, Antov A, Jankovic D, Sher A, Radtke F, Souabni A, et al. Direct regulation of Gata3 expression determines the T helper differentiation potential of Notch. Immunity. 2007;27:89–99.CrossRefPubMedCentralPubMed
47.
Zurück zum Zitat Wang J, Lee S, Teh CE, Bunting K, Ma L, Shannon MF. The transcription repressor, ZEB1, cooperates with CtBP2 and HDAC1 to suppress IL-2 gene activation in T cells. Int Immunol. 2009;21:227–35.CrossRefPubMed Wang J, Lee S, Teh CE, Bunting K, Ma L, Shannon MF. The transcription repressor, ZEB1, cooperates with CtBP2 and HDAC1 to suppress IL-2 gene activation in T cells. Int Immunol. 2009;21:227–35.CrossRefPubMed
Metadaten
Titel
CD4 T-cell transcriptome analysis reveals aberrant regulation of STAT3 and Wnt signaling pathways in rheumatoid arthritis: evidence from a case–control study
verfasst von
Hua Ye
Jing Zhang
Jun Wang
Yanyan Gao
Yan Du
Chun Li
Minghua Deng
Jianping Guo
Zhanguo Li
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 1/2015
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-015-0590-9

Weitere Artikel der Ausgabe 1/2015

Arthritis Research & Therapy 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Wo hapert es noch bei der Umsetzung der POMGAT-Leitlinie?

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Das Risiko für Vorhofflimmern in der Bevölkerung steigt

02.05.2024 Vorhofflimmern Nachrichten

Das Risiko, im Lauf des Lebens an Vorhofflimmern zu erkranken, ist in den vergangenen 20 Jahren gestiegen: Laut dänischen Zahlen wird es drei von zehn Personen treffen. Das hat Folgen weit über die Schlaganfallgefährdung hinaus.

VHF-Ablation nützt wohl nur bei reduzierter Auswurfleistung

02.05.2024 Ablationstherapie Nachrichten

Ob die Katheterablation von Vorhofflimmern bei Patienten mit Herzinsuffizienz die Komplikationsraten senkt, scheint davon abzuhängen, ob die Auswurfleistung erhalten ist oder nicht. Das legen die Ergebnisse einer Metaanalyse nahe.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.