Open Access

Differences in atrial fibrillation‑associated proteins between the left and right atrial appendages from patients with rheumatic mitral valve disease: A comparative proteomic analysis

Corrigendum in: /10.3892/mmr.2018.9218

  • Authors:
    • Hai Liu
    • Guangxian Chen
    • Hongsheng Zheng
    • Han Qin
    • Mengya Liang
    • Kangni Feng
    • Zhongkai Wu
  • View Affiliations

  • Published online on: September 26, 2016     https://doi.org/10.3892/mmr.2016.5776
  • Pages: 4232-4242
  • Copyright: © Liu 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

The majority of proteomic studies have focused on identifying atrial fibrillation (AF)-associated proteins in the right atrium (RA), thus potential differences in AF‑associated proteins between the RA and left atrium (LA) remain unknown. The aim of the present study was to perform proteomic analysis to compare the potential differences in AF‑associated proteins between the right atrial appendage (RAA) and left atrial appendage (LAA) in patients with rheumatic mitral valve disease (RMVD). RAA and LAA tissues were obtained from 18 patients with RMVD (10 with AF) during mitral valve replacement surgery. Two‑dimensional fluorescence difference gel electrophoresis (2‑D DIGE) proteomics analysis was performed using these tissues to identify AF‑associated proteins in RAA and LAA. Subsequently, the proteomics data was validated using western blot analysis of nine selected proteins. In RAA, 32 AF‑associated proteins were significantly dysregulated (15 upregulated and 17 downregulated). In LAA, 31 AF‑associated proteins were significantly dysregulated (13 upregulated and 18 downregulated). Among these AF‑associated proteins, 17 were AF‑associated in both RAA and LAA, 15 were AF‑associated only in RAA, and 14 were AF‑associated only in LAA. Amongst the differentially expressed proteins, western blot analysis validated the results for 6 AF‑associated proteins, and demonstrated similar distributions in RAA and LAA compared with the 2‑D DIGE results. Of these proteins, 2 proteins were AF‑associated in both RAA and LAA, 2 were AF‑associated only in RAA, and 2 were AF‑associated only in LAA. Additionally, the different distributions of AF‑associated proteins in the RAA and LAA of patients with RMVD was analyzed, which may reflect the different regulatory mechanisms of the RA and LA in AF. These findings may provide new insights into the underlying molecular mechanisms of AF in patients with RMVD.

Introduction

Atrial fibrillation (AF) is the most common sustained arrhythmia in clinical practice (1). It often occurs concomitantly with other cardiovascular diseases, including hypertension, congestive heart failure, coronary artery disease, and valvular heart disease (2,3). Rheumatic mitral valve disease (RMVD), a major cardiovascular disease in developing countries affected by rheumatic fever, is a major clinical risk factor for AF (4,5). Development of effective therapies and preventative strategies are crucial for the control of AF-associated morbidity and mortality. However, Currently, medical interventions for AF are relatively limited, as the precise mechanisms of AF have not been completely elucidated. Therefore, novel methods to probe the underlying mechanisms of AF and potential novel mechanism-based therapeutic strategies are required (6).

The field of cardiovascular proteomics has shed new light on understanding the regulatory mechanisms of AF (7). Progression from paroxysmal AF to persistent, and then to permanent AF involves complex changes in gene expression, and subsequent changes in protein expression and activity (8). Proteomic techniques are a powerful tool to evaluate global protein changes in diseased hearts and discover novel proteins, diagnostic biomarkers, and potential drug targets for the development of novel therapeutic agents (7). Two-dimensional fluorescence difference gel electrophoresis (2-D DIGE) can provide an overview of the proteome during disease (9). Previous use of proteomic methods have identified differentially expressed proteins associated with AF (AF-associated proteins) in several animal models (10,11) and human heart tissue (1216).

Genomic (8,17), morphological (18,19), and electrophysiological (2022) differences have been observed between the right atrium (RA) and left atrium (LA). This suggests that different mechanisms regulate the RA and LA in AF (23). Thus, it is unsurprising that the AF-associated proteins of the RA may differ from those of the LA. However, the majority AF proteomics studies have primarily focused on the RA. Thus, potential differences in AF-associated proteins between RA and LA remain unknown. Given the complexity of AF progression, a better understanding of the differences in AF-associated proteins between RA and LA may elucidate novel cardioprotective strategies.

Thus, the aim of the present study was to perform 2-D DIGE proteomics analysis to compare potential differences of AF-associated proteins in the right atrial appendage (RAA) and left atrial appendage (LAA) from patients with RMVD that were either in sinus rhythm (SR) or AF.

Materials and methods

Ethics approval

Approval for this study was obtained from the Human Ethics Committee at the First Affiliated Hospital of Sun Yat-Sen University. The investigation complied with the principles that govern the use of human tissues outlined in the Declaration of Helsinki. All patients provided informed consent prior to participation in the study.

Human tissue preparation

Human tissue preparation was performed as previously described (8). Briefly, RAA and LAA tissues were obtained from the same patient as surgical biopsies at the time of the mitral valve replacement surgery (RAA from cannulation site and LAA during ligation). Tissues were immediately snap frozen in liquid nitrogen, and stored at −80°C prior to use. Tissue samples from the RAA and LAA were obtained from 18 patients with RMVD. The patients in the SR group (n=8) had no history of AF. The patients in the AF group (n=10) presented with a documented history of arrhythmias for >6 months prior to surgery. The diagnosis of AF was reached by evaluating medical records and 12-lead electrocardiogram findings. Patients with SR also had no history of using antiarrhythmic drugs and were screened to ensure that they had never experienced AF. Routine pre-operative color Doppler echocardiography was performed on all patients. Pre-operative functional status was recorded according to New York Heart Association (NYHA) classifications.

Preparation of protein extracts

Protein extracts were isolated from tissues based on previously published protocols (24). Briefly, tissues (approximately 0.1 g) were homogenized in lysis buffer [7 M urea, 2 M thiourea, 30 mM Tris, 4% (w/v) CHAPS cell lysis buffer, 40 mM dithiothreitol (DTT), 0.6 mM phenylmethylsulfonyl fluoride] and the supernatant was collected following centrifugation (2,000 × g, 4°C, 40 min). To remove non-protein material from the extract and determine the final protein concentration, the 2-D Clean-up kit (GE Healthcare Life Sciences, Chalfont, UK) and 2-D Quant kit (GE Healthcare Life Sciences) were used according to manufacturer's instructions.

Protein labeling with CyDye DIGE fluorophores

Protein extracts were labeled with three CyDye DIGE fluorophores, Cy2, Cy3 and Cy5 (GE Healthcare Life Sciences), for 2-D DIGE technology, according to the manufacturer's protocols. Briefly, equal amounts of protein from the same group were pooled and divided into four equal portions (50 µg each). The latter portions were labeled with 400 pmol Cy3 or Cy5 according to the experimental design. A pooled sample consisting of equal amounts of all samples was used as the pooled internal standard and labeled with Cy2. After incubating samples on ice for 30 min in the dark, the labeling reaction was stopped with 10 mM lysine. For each gel, Cy2-, Cy3- and Cy5-labeled proteins (50 µg each) were mixed and calibrated to 450 µl with rehydration buffer [7 M urea, 2 M thiourea, 4% (w/v) CHAPS cell lysis buffer, 40 mM DTT, 1% IPG buffer (pH 4–7), 0.002% (w/v) bromophenol blue].

2-D electrophoresis

2-D electrophoresis was performed as previously described (25). The labeled protein mixture in each gel was applied to ImmobilineDryStrip strips (24 cm, pH 4–7; GE Healthcare Life Sciences). Isoelectric focusing (IEF) was performed with an EttanIPGphor II apparatus (GE Healthcare Life Sciences) using the follow steps: 30 V for 12 h, 500 V for 1 h, 1,000 V for 1 h, and 10,000 V for up to a total of 85,000 V/h. Following IEF, the proteins were reduced and alkylated by successive 15 min treatments with equilibration buffer containing 2% (w/v) DTT followed by 2.5% (w/v) iodoacetamide. Proteins were then resolved on 12.5% SDS-PAGE gels using an EttanDALTsix instrument (GE Healthcare Life Sciences). In order to facilitate mass spectrometry analysis, 500 µg of unlabeled pool protein sample was run in parallel on a preparative gel and stained by Deep Purple staining (GE Healthcare Life Sciences) according to the manufacturer's instructions.

Gel image acquisition and analysis

Gel images were acquired on a Typhoon 9400 scanner (GE Healthcare Life Sciences) and analyzed using DeCyder software (version 6.0; GE Healthcare Life Sciences) as previously described (26,27). Briefly, Cy2-, Cy3- and Cy5-labeled images of each gel were scanned with excitation/emission wavelengths of 488/520, 532/580 and 633/670 nm, respectively. Following CyDye labeling, signals were imaged and the gels were stained using Deep Purple total protein stain (GE Healthcare Life Sciences) according to standard protocols and scanned with excitation/emission wavelengths of 532/560 nm. The 2-D DIGE gel images were subsequently analyzed with DeCyder software. Protein expression patterns for SR-RAA were compared with AF-RAA, and SR-LAA were compared with AF-LAA. Ratios of proteins that increased or decreased >1.5-fold (t-test, P<0.05) were considered significant changes (9). The corresponding protein spots were also selected in the stained preparative gel for spot picking.

Spot picking and enzymatic digestion

Selected protein spots in the preparative gels were automatically picked and handled in an Ettan Spot Handling Workstation (GE Healthcare Life Sciences) for the preparation of the protein sample for matrix-assisted laser desorption/ionization time-of-flight mass spectrometer/mass spectrometry (MALDI-TOF/TOF MS) analysis. In the automated procedure, the selected protein spots were picked, washed with 15 mM NH4HCO3 and 50% v/v methanol, then digested with 20 ng/ml trypsin (sequencing grade; Promega Corporation, Madison, WI, USA) in 20 mM NH4HCO3 for 2 h at 37°C. Tryptic peptides were extracted with 50% v/v acetonitrile (ACN) and 0.5% v/v trifluoroacetic acid (TFA), and dissolved in 5 mg/ml R-cyano-4-hydroxycinnamic acid (GE Healthcare Life Sciences) matrix in 50% (v/v) ACN and 0.1% (v/v) TFA. Finally, samples were spotted on the mass spectrometry sample plate.

MALDI-TOF/TOF MS analysis and database searching

Mass spectrometry analysis was performed using an Utraflex III MALDI-TOF/TOF MS (Bruker Corporation, Ettlingen, Germany) operating in a positive ion reflector mode. Monoisotopic peak masses were acquired in a mass range of 700–4,000 Da, with a signal-to-noise ratio >200. Four of the most intense ion signals, excluding common trypsin autolysis peaks and matrix ion signals, were automatically selected as precursors for mass spectrometer/mass spectrometry (MS/MS) acquisition. Peptide mass fingerprint (PMF) combined with MS/MS spectra was blasted against the NCBInr database using the Biotools software (version 3.2; Bruker Corporation) and MASCOT version 2.2 (Matrix Science, Inc., Boston, MA, USA). Search parameters were set as follows: Homo sapiens, trypsin cleavage, one missed cleavage allowed, carbamidomethylation as fixed modification, oxidation of methionines allowed as variable modification, peptide mass tolerance at 50 ppm, and fragment tolerance at 0.5 Da. The criteria for successfully identifying a protein was as follows: Ion score confidence interval (%) for PMF and MS/MS data >95%, peptide count (hit) ≥4, and at least two peptides of distinct sequence were identified in MS/MS analysis.

Protein categorization and protein-protein interaction prediction

Protein categorization and protein-protein interaction predictions were performed as previously described (9). Briefly, the identified proteins were classified, according to the Protein Analysis Through Evolutionary Relationships (PANTHER) system (http://www.pantherdb.org), which classified genes and proteins by their function. The PANTHER ontology, a highly controlled vocabulary (ontology terms) by molecular function, biological processes and molecular pathways, was employed to categorize proteins into families and subfamilies with shared function. The prediction of protein-protein interactions was performed by utilizing the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) system (http://www.string-db.org). STRING is a database of known and predicted protein interactions that include direct (physical) and indirect (functional) associations.

Western blot analysis

Western blot analysis was performed as previously described (28). Briefly, protein lysates were prepared from human tissues, separated on SDS-PAGE, transferred to a polyvinylidene difluoride membrane, and subjected to immunoblotting at 4°C overnight with the following antibodies: Anti-aldo-keto reductase family 1 member B10 (AKR1B10; diluted 1:10,000; polyclonal antibody; cat. no. S1646; Epitomics, Burlingame, CA, USA), anti-crystallin α B (CRYAB; diluted 1:2000; polyclonal antibody; cat. no. PAB7394; Abnova Corporation, Taipei, Taiwan), anti-annexin 4 (ANAX4; diluted 1:800; polyclonal antibody; cat. no. H00000307-B01P; Abnova Corporation), anti-G protein subunit α o1 (GNAO1; diluted 1:10,000; polyclonal antibody; cat. no. S3107; Epitomics), anti-ribonuclease H1 (RNase H1; diluted 1:800; monoclonal antibody; cat. no. ab56560; Abcam, Cambridge, MA, USA), anti-moesin (MSN; diluted 1:2,000; polyclonal antibody; cat. no. PAB7062; Abnova Corporation), anti-nestin (NES; diluted 1:6,000; polyclonal antibody; cat. no. PAB12375; Abnova Corporation) and anti-transferrin (diluted 1:8,000; polyclonal antibody; cat. no. S0860; Epitomics), anti-osteoglycin (diluted 1:500; monoclonal antibody; cat. no. 5518-1; Epitomics). β-actin was assessed as a loading control (diluted 1:20,000; monoclonal antibody; cat. no. 60008-1-Ig; ProteinTech Group, Inc., Chicago, IL, USA). After washing, membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies (goat anti-mouse, cat. no. SA00001-1; goat anti-rabbit, cat. no. SA00001-2; or rabbit anti-goat, cat. no. SA00001-4; all from ProteinTech Group, Inc.) and visualized using an enhanced chemiluminescence western blot detection system (Immobilon Western Chemiluminescent HRP substrate; Merck Millipore, Darmstadt, Germany) on X-ray film (Kodak, Rochester, NY, USA). All the proteins were detected on the same blot. For certain protein with molecular weights very close to each other, the blots were stripped and reprobed using Restore™ PLUS Western Blot Stripping Buffer(Thermo Fisher Scientific Inc., Waltham, MA, USA) according to the manufacturer's instructions. Images were scanned and analyzed using Kodak Molecular Imaging software (version 5.0). Non-specific binding was subtracted, and the band signals were expressed as relative protein amounts compared with β-actin protein levels.

Statistical analysis

Data of clinical characteristics of patients and western blot quantification are presented as the mean ± standard deviation. Student's t-test was used for statistical comparison of the data and to calculate significant differences in the relative abundance of individual protein spots between the two groups during 2-D DIGE analysis. The SPSS 16.0 software package (SPSS, Inc., Chicago, IL, USA) was used to conduct the statistical analyses and P<0.05 (two-tailed) was considered to indicate a statistically significant difference.

Results

Clinical characteristics of the patients with SR and AF

Tissue from RAA and LAA was obtained from each patient. No significant differences in terms of age, gender or NYHA class were observed between the SR and AF groups. Pre-operative color Doppler echocardiography demonstrated that the LA size of the patients with AF was significantly increased compared with the SR patients. However, no differences in the left ventricular end-diastolic diameter and left ventricular ejection fraction were observed between the groups (Table I).

Table I.

Clinical characteristics of the patients with SR and AF.

Table I.

Clinical characteristics of the patients with SR and AF.

CharacteristicSR (n=8)AF (n=10)
Gender (male/female)5/35/5
Age (years)50.16±6.8851.42±7.12
LA size (mm)43.31±3.23 57.65±5.08a
LVEDD (mm)48.35±5.1550.13±3.07
LVEF (%)61.34±5.6658.71±3.81
NYHA class
  II67
  III23

a P<0.05 vs. SR patients. Values are presented as the mean ± standard deviation. SR, sinus rhythm; AF, atrial fibrillation; LA, left atrium; LVEDD, left ventricular end-diastolic diameter; LVEF, left ventricular ejection fraction; NYHA class, New York Heart Association classifications.

2-D DIGE analysis and MALDI-TOF/TOF MS identification

Following 2-D DIGE analysis, a total of 2,813 spots were observed to be well matched across all gels using the DeCyder software analysis. The 2-D DIGE gel images are presented in Fig. 1. Following visual review, 61 protein spots of high abundance that exhibited significantly altered expression (AF-RAA vs. SR-RAA and AF-LAA vs. SR-LAA) were selected for MALDI-TOF/TOF MS analysis. A total of 32 differentially expressed proteins were identified, including 15 that were upregulated and 17 that were downregulated in the AF-RAA tissues compared with the SR-RAA tissues (Table II). Additionally, a total of 31 differentially expressed proteins, including 13 that were upregulated and 18 that were downregulated in the AF-LAA tissues compared with the SR-LAA tissues (Table II).

Table II.

Differential proteins identified by mass spectrometry following 2-D DIGE of the RAA and LAA samples.

Table II.

Differential proteins identified by mass spectrometry following 2-D DIGE of the RAA and LAA samples.

Av. ratiobt-test
Accession no.aMW (kDa)pIRAAcLAAdRAAcLAAdProtein name
In RAA and LAA (n=17)
  gi|886992.411.551.72.360.00650.013T-cell receptor β chain J region (clone HBP22)-human (fragment)
  gi|76695501245.51.561.50.00120.0044Vinculin isoform meta-VCL
  gi|5669804729.1−1.76−1.830.0160.00039Polycystin-2-like protein
  gi|42543698426.621.571.50.0380.011Chain A, The Crystal Structure Of The Human Hsp70 Atpase Domain
  gi|413509231105.851.612.060.0110.0017Collagen, type VI, α 2
  gi|386973615.3−1.88−1.870.0220.00066β-Myosin heavy chain
  gi|386972625.3−1.88−1.910.010.0019α-Myosin heavy chain
  gi|386970715.32−1.79−1.940.0190.00019Myosin heavy chain β-subunit
  gi|33150528e345.33−1.75−2.010.0120.0079Osteoglycin
  gi|223468663e367.67−1.68−2.020.0260.0026Aldo-ketoreductase family 1 member B10
  gi|19908424545.211.991.940.0360.038Mutant desmin
  gi|189617e365.651.551.730.0210.026Protein PP4-X
  gi|1196223668.010.91−2.23−2.080.0180.00011hCG1813095
  gi|1196220461612.02−1.58−1.680.00190.023hCG1646228
  gi|119612724304.88−1.79−2.020.0140.0028Actin, α, cardiac muscle, isoform CRA_c
  gi|1195865592245.58−2.57−2.140.00820.0052Myosin, heavy polypeptide 6, cardiac muscle, α (cardiomyopathy, hypertrophic 1), isoform CRA_b
  gi|119576987349.33−1.88−2.030.0110.0041DNA cross-link repair 1B
Only in RAA (n=15)
  gi|94981553194.92−2.720.024Myosin regulatory light chain 2, ventricular/cardiac muscle isoform
  gi|62897129715.28−1.550.027Heat shock 70kDa protein 8 isoform 1 variant
  gi|55749932545.212.040.0035Desmin
  gi|51103391128.98−1.680.011Immunoglobulin variable region VL kappa domain
  gi|5031875656.41.60.017Lamin-A/C isoform 2
  gi|46593007535.944.820.048Cytochrome b-c1 complex subunit 1, mitochondrial precursor
  gi|4505257e686.081.620.029Moesin
  gi|4505047396.16−1.630.023Lumican precursor
  gi|4503057e206.761.990.0016α-Crystallin B chain
  gi|31615330685.662.090.0091Chain A, human serum albumin mutant R218h complexed with thyroxine
  gi|223170475.541.560.032Fibrinogen γ
  gi|1459423339.65.51.790.019Immunoglobulin heavy chain variable region
  gi|119581148586.95−1.690.002Keratin 9
  gi|116283748e665.15−2.040.0068Nestin protein
  gi|114615454446.571.790.015PREDICTED: dihydrolipoamide dehydrogenase isoform 1
Only in LAA (n=14)
  gi|9984673.14.411.570.04448 kDa histamine receptor subunit peptide 4
  gi|62414289545.062.710.0035Vimentin
  gi|56204818385.39−1.840.0016Actin, α 1, skeletal muscle
  gi|4885079339.312.230.0085ATP synthase subunit γ, mitochondrial isoform H (heart) precursor
  gi|484769731375.29−1.670.0047Rhabdomyosarcoma antigen MU-RMS-40.7B
  gi|45018931045.312.170.0078α-Actinin-2
  gi|348113701310.15−2.260.0062Chain B, crystal structure of the 46kDa domain of human cardiac troponin in the Ca2+ saturated form
  gi|34190601e355.18−1.690.00024G protein subunit α o1 protein
  gi|220806729.610.961.60.00089MHC class I antigen
  gi|15029922e504.83−1.670.011Ribonuclease H1 protein
  gi|1196185381310.951.760.0086hCG2016250, isoform CRA_g
  gi|119612436368.27−1.970.0002hCG1779566, isoform CRA_a
  gi|110590599e776.851.580.014Chain A, apo-human serum transferrin
  gi|109102505355.82−1.60.0024PREDICTED: similar to serine/threonine-protein phosphatase PP1-β catalytic subunit (PP-1B) isoform 1

a Accession number of NCBInr database.

b Decreased or increased ratio of AF-RAA comparison with SR-RAA, and AF-LAA comparison with SR-LAA.

c AF-RAA comparison to SR-RAA.

d AF-LAA comparison with SR-LAA.

e Selected for validation by western blotting. “-” indicates, not detected. AF, atrial fibrillation; SR, sinus rhythm; MW, molecular weight; RAA, right atrial appendage; LAA, left with appendage.

Comparison of differentially expressed proteins between RAA and LAA tissues

Using 2-D DIGE proteomics methodology, a total of 46 differentially expressed proteins were detected (in RAA or LAA tissues). Among these, 32 proteins were expressed in RAA tissues, and 31 proteins in LAA tissues. Furthermore, 17 proteins were identified in RAA and LAA tissues, whereas 15 proteins were detected only in RAA tissues and 14 proteins were only in LAA tissues (Table II; Fig. 2).

Classification of differentially expressed proteins and the protein-protein interaction network

According to cellular function and processes, a total of 46 differentially expressed proteins were distributed into categories based on molecular function, biological processes and protein classes using the PANTHER classification system. Results are presented in Fig. 3. The most dominant function that the identified proteins were involved in was structural activity (34.8%), followed by binding (19.6%), catalytic activity (19.6%), transporter activity (8.7%), receptor activity (8.7%), nucleic acid/protein binding transcription factor activity (4.4%) and enzyme regulator activity (4.3%). The majority of the identified proteins had functions in cellular processes (17.3%), cellular component organization or biogenesis (13.6%), metabolic processes (13.6%) and developmental processes (13.6%). Other biological processes that the proteins were involved in included localization (9.1%), response to stimuli (9.1%), and multicellular organismal processes (7.3%). As for protein class, the majority of the proteins belonged in categories associated with cytoskeletal proteins (23.7%) and structural proteins (10.2%), while others belonged to categories associated with enzyme modulators (6.8%), receptors (6.8%) and calcium-binding proteins (3.4%).

The protein interaction network for the identified proteins was constructed by STRING (Fig. 4) in order to improve the understanding of the pathogenic mechanisms in AF with RMVD. Biological systems can be modeled as complex network systems with many interactions between components of different pathways. These interactions provide us with essential information about the function and behavior of the analyzed proteins (9).

Validation of the 2-D DIGE proteomics data by western blot analysis

To validate the data obtained from the 2-D DIGE proteomics, western blot analysis was performed on 9 selected proteins (Table II). According to the western blot data, AKR1B10 and ANAX4 were AF-associated proteins in RAA and LAA tissues (Fig. 5), whereas CRYAB and MSN were AF-associated proteins in only RAA tissues. RNase H1 and transferrin were AF-associated proteins only in LAA tissues (Fig. 5). The western blot data validated the results for all of these proteins, excluding osteoglycin, NES and GNAO1, AF-associated proteins which were not detected to be differentially expressed in the RAA or LAA of AF tissues when assessed using western blot analysis (Fig. 5).

Comparative analysis of the expression of six validated proteins between the RAA and LAA tissues from RMVD patients with SR

Six differentially expressed proteins were identified and validated using 2-D DIGE proteomics and western blot analysis. These proteins had different tissue distributions between RAA and LAA. Additionally, it was aimed to elucidate whether expression of these proteins was also different between RAA and LAA tissues based on SR status. Western blot analysis was used to compare the expression level of these proteins between the SR-RAA and SR-LAA groups. Expression levels of AKR1B10, ANAX4 and RNase H1 were not different in the RAA and LAA of patients with SR (Fig. 6). However, CRYAB and MSN expression was increased in the SR-LAA group compared with SR-RAA (Fig. 6). Transferrin expression was increased in the SR-RAA group compared with the SR-LAA group (Fig. 6).

Discussion

Previous studies have identified that both the RA (29,30) and LA (31,32) serve an important role in AF. However, intervention strategies that only target the RA or LA do not ameliorate all of the complications associated with AF (3335). This may reflect the necessity to understand the different mechanisms involved in AF between the RA and LA. The development of AF involves complex changes in gene expression and subsequent changes in protein expression and activity. Thus, it is important to understand the protein mechanisms of AF to facilitate the development of potential novel mechanism-based therapeutic strategies. Proteomic technology provides a comprehensive strategy to investigate AF-associated proteins. Recently, there have been many proteomic studies associated with AF (1016). However, the majority of proteomics studies on AF primarily focus on the RA and less on the LA. Although two previous studies (12,16) used proteomics analysis to identify AF-associated proteins in RA and LA tissue from patients with mitral valve disease, potential differences in AF-associated proteins between the RA and LA were not determined. Therefore, the potential differential expression of AF-associated proteins between the RA and LA remain unknown. To the best of our knowledge, the current study is the first to compare the potential differences of AF-associated proteins in the RA and LA from RMVD patients.

Profiling protein expression patterns at specific stages of disease can reflect the status of disease progression. We hypothesize that if there are different mechanisms involved in AF between the RA and LA, then changes in protein expression profiles will also reflect this pattern. The differentially expressed proteins were compared between RA and LA to reveal different disease mechanisms underlying AF in the RA and LA. The present study demonstrated that the development of AF in patients with RMVD was associated with significant changes in protein expression in RAA and LAA tissues and that these AF-associated proteins had different distributions in RAA and LAA. Certain AF-associated proteins were differentially expressed both in the RA and LA compared with expression in SR samples, while others were different only in the RA or LA. The current study further demonstrated that the expression of certain proteins were not changed between the RAA and LAA in patients with SR, whereas there is a difference in the protein expression profiles of SR and AF between the RAA and LAA. Therefore, it was hypothesize that differential distribution of these AF-associated proteins may reflect different protein mechanisms in the RA and LA during AF. Genomic (8,17), morphological (18,19), and electrophysiological (2022) differences have been observed between the RA and LA in mechanisms involved in AF. Thus, the present study provided proteomic evidence that different mechanisms regulate the RA and LA in AF.

Protein expression profiles also reflect certain spatial characteristics (depending on cell, tissue or organ type). The current study observed that the protein expression levels of CRYAB, MSN and transferrin were different when comparing RAA and LAA tissues in patients with RMVD and SR. CRYAB and MSN expression was increased in the SR-LAA group compared with the SR-RAA group, however they were identified as AF-associated proteins only in RAA tissue. By contrast, transferrin expression was increased in the SR-RAA group compared with the SR-LAA, and was identified as an AF-associated protein only in LAA tissue. These results suggest that CRYAB, MSN and transferrin are tissue dependent AF-associated proteins that may be involved in the progression from SR to AF as observed by the initial expression studies performed in RAA and LAA of RMVD patients with SR. The difference in the initial expression of these proteins between RAA and LAA in patients with RMVD and SR may due to the progress from healthy to RMVD status. Cooley et al (36) reported that microRNA expression profiles caused differential changes in the RA and LA with the development from healthy to valvular heart diseased condition. Thus, potentially, the certain proteins become differentially expressed in the RA and LA during RMVD compared with healthy settings. In addition, in patients with RMVD, the association between LA size and AF is well established and LA dilatation is considered to be a cause and a consequence of AF (5). The results of the present study revealed that the LA size of patients with AF was significantly greater than in patients with SR (Table I), thus, potentially, the significant structural remodeling occurring in the LA may also alter the protein expression profiles and cause, at least partially, differential regulation of AF-associated proteins in the RA and LA in patients with RMVD.

The current study identified several AF-associated proteins using 2-D DIGE proteomics analysis. Of these proteins, a number may participate in the mechanisms associated with AF, whereas others maybe a result of AF. The exact role of these differentially expressed proteins in AF requires further investigation. Stringent bioinformatics analysis will be required to select for candidate proteins for future functional studies. Bioinformatics analysis was performed in the current study using the PANTHER and STRING systems to classify AF-associated proteins and predict a protein-protein interaction network (Figs. 5 and 6). This analysis may provide investigators with vital information to direct future research.

A major limitation of this study was the small number of patients. This was due, in part, to the difficulty in finding patients with RMVD and SR. In addition, because the study was performed with human tissues with existing disease, experiments could not be conducted to modulate the protein levels. Therefore, exact targets and pathways by which alterations in these proteins may cause AF in patients with RMVD remain elusive and require further investigation. Finally, patients in this study were a specific cohort with preserved systolic left ventricular function and little comorbidity, undergoing mitral valve replacement surgery. Thus, changes identified in this population may not be representative of other cohort populations.

In conclusion, the current study identified differentially expressed proteins that have potential associations with AF in the RAA and LAA tissues from patients with RMVD. The different distribution of these AF-associated proteins may reflect different mechanisms underlying RAA and LAA involvement in AF. These findings may be useful for the biological understanding of AF in patients with RMVD and suggest potential therapeutic targets for AF.

Acknowledgements

This work is supported by the Pearl River Scholar Program (grant no. 80000-3210003) and the National Natural Science Foundation of China (grant nos. 81370215 and 81500260). The authors wish to acknowledge Dr. Kun-hua Hu (Key Laboratory on Proteomics, Sun Yat-sen University, Guangzhou, China) for his technical expertise. We are grateful to Dr. Gang Dai and Dr. Rong Fang (Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China) for their technical expertise.

Glossary

Abbreviations

Abbreviations:

AF

atrial fibrillation

SR

sinus rhythm

RMVD

rheumatic mitral valve disease

RA

right atrium

LA

left atrium

RAA

right atrial appendage

LAA

left atrial appendage

2-D DIGE

two-dimensional fluorescence difference gel electrophoresis

IEF

isoelectric focusing

MALDI-TOF/TOF MS

matrix-assisted laser desorption/ionization time-of-flight mass spectrometer/mass spectrometry

MS/MS

mass spectrometer/mass spectrometry

PMF

peptide mass fingerprint

References

1 

Murphy NF, Simpson CR, Jhund PS, Stewart S, Kirkpatrick M, Chalmers J, MacIntyre K and McMurray JJ: A national survey of the prevalence, incidence, primary care burden and treatment of atrial fibrillation in Scotland. Heart. 93:606–612. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Darby AE and Dimarco JP: Management of atrial fibrillation in patients with structural heart disease. Circulation. 125:945–957. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Rosiak M, Dziuba M, Chudzik M, Cygankiewicz I, Bartczak K, Drozdz J and Wranicz JK: Risk factors for atrial fibrillation: Not always severe heart disease, not always so ‘lonely’. Cardiol J. 17:437–442. 2010.PubMed/NCBI

4 

Benjamin EJ, Levy D, Vaziri SM, D'Agostino RB, Belanger AJ and Wolf PA: Independent risk factors for atrial fibrillation in a population-based cohort. The Framingham Heart Study. JAMA. 271:840–844. 1994. View Article : Google Scholar : PubMed/NCBI

5 

Carabello BA: Modern management of mitral stenosis. Circulation. 112:432–437. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Nattel S: New ideas about atrial fibrillation 50 years on. Nature. 415:219–226. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Lam L, Lind J and Semsarian C: Application of proteomics in cardiovascular medicine. Int J Cardiol. 108:12–19. 2006. View Article : Google Scholar : PubMed/NCBI

8 

Liu H, Qin H, Chen GX, Liang MY, Rong J, Yao JP and Wu ZK: Comparative expression profiles of microRNA in left and right atrial appendages from patients with rheumatic mitral valve disease exhibiting sinus rhythm or atrial fibrillation. J Transl Med. 12:902014. View Article : Google Scholar : PubMed/NCBI

9 

Deng L, Jia HL, Liu CW, Hu KH, Yin GQ, Ye JW, He CH, Chen JH, Xie YP, Dang R, et al: Analysis of differentially expressed proteins involved in hand, foot and mouth disease and normal sera. Clin Microbiol Infect. 18:E188–E196. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Lai LP, Lin JL, Lin CS, Yeh HM, Tsay YG, Lee CF, Lee HH, Chang ZF, Hwang JJ, Su MJ, et al: Functional genomic study on atrial fibrillation using cDNA microarray and two-dimensional protein electrophoresis techniques and identification of the myosin regulatory light chain isoform reprogramming in atrial fibrillation. J Cardiovasc Electrophysiol. 15:214–223. 2004. View Article : Google Scholar : PubMed/NCBI

11 

De Souza AI, Cardin S, Wait R, Chung YL, Vijayakumar M, Maguy A, Camm AJ and Nattel S: Proteomic and metabolomic analysis of atrial profibrillatory remodelling in congestive heart failure. J Mol Cell Cardiol. 49:851–863. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Modrego J, Maroto L, Tamargo J, Azcona L, Mateos-Cáceres P, Segura A, Moreno-Herrero R, Pérez-Castellanos N, Delpón E, Pérez-Villacastín J, et al: Comparative expression of proteins in left and right atrial appendages from patients with mitral valve disease at sinus rhythm and atrial fibrillation. J Cardiovasc Electrophysiol. 21:859–868. 2010.PubMed/NCBI

13 

Mayr M, Yusuf S, Weir G, Chung YL, Mayr U, Yin X, Ladroue C, Madhu B, Roberts N, De Souza A, et al: Combined metabolomic and proteomic analysis of human atrial fibrillation. J Am Coll Cardiol. 51:585–594. 2008. View Article : Google Scholar : PubMed/NCBI

14 

García A, Eiras S, Parguiña AF, Alonso J, Rosa I, Salgado-Somoza A, Rico TY, Teijeira-Fernández E and González-Juanatey JR: High-resolution two-dimensional gel electrophoresis analysis of atrial tissue proteome reveals down-regulation of fibulin-1 in atrial fibrillation. Int J Cardiol. 150:283–290. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Huang WJ, Zhou R, Zeng XR, Tan XQ, Cheng ZH, Tang MH, Gou LT, Chen LJ, Tong AP, He Y and Yang JL: Comparative proteomic analysis of atrial appendages from rheumatic heart disease patients with sinus rhythm and atrial fibrillation. Mol Med Rep. 4:655–661. 2011.PubMed/NCBI

16 

Zhang P, Wang W, Wang X, Wang X, Song Y, Han Y, Zhang J and Zhao H: Protein analysis of atrial fibrosis via label-free proteomics in chronic atrial fibrillation patients with mitral valve disease. PLoS One. 8:e602102013. View Article : Google Scholar : PubMed/NCBI

17 

Zhu H, Zhang W, Zhong M, Zhang G and Zhang Y: Differential gene expression during atrial structural remodeling in human left and right atrial appendages in atrial fibrillation. Acta Biochim Biophys Sin (Shanghai). 43:535–541. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Park JH, Lee JS, Ko YG, Lee SH, Lee BS, Kang SM, Chang BC and Pak HN: Histological and biochemical comparisons between right atrium and left atrium in patients with mitral valvular atrial fibrillation. Korean Circ J. 44:233–242. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Embi AA, Scherlag BJ and Ritchey JW: Glycogen and the propensity for atrial fibrillation: Intrinsic anatomic differences in glycogen in the left and right atria in the goat heart. N Am J Med Sci. 6:510–515. 2014. View Article : Google Scholar : PubMed/NCBI

20 

John B, Stiles MK, Kuklik P, Chandy ST, Young GD, Mackenzie L, Szumowski L, Joseph G, Jose J, Worthley SG, et al: Electrical remodelling of the left and right atria due to rheumatic mitral stenosis. Eur Heart J. 29:2234–2243. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Tai CT, Lo LW, Lin YJ and Chen SA: Arrhythmogenic difference between the left and right atria in a canine ventricular pacing-induced heart failure model of atrial fibrillation. Pacing Clin Electrophysiol. 35:188–195. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Li D, Zhang L, Kneller J and Nattel S: Potential ionic mechanism for repolarization differences between canine right and left atrium. Circ Res. 88:1168–1175. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Chen J, Mandapati R, Berenfeld O, Skanes AC, Gray RA and Jalife J: Dynamics of wavelets and their role in atrial fibrillation in the isolated sheep heart. Cardiovasc Res. 48:220–232. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Liu W, Zhou XW, Liu S, Hu K, Wang C, He Q and Li M: Calpain-truncated CRMP-3 and −4 contribute to potassium deprivation-induced apoptosis of cerebellar granule neurons. Proteomics. 9:3712–3728. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Pang J, Liu WP, Liu XP, Li LY, Fang YQ, Sun QP, Liu SJ, Li MT, Su ZL and Gao X: Profiling protein markers associated with lymph node metastasis in prostate cancer by DIGE-based proteomics analysis. J Proteome Res. 9:216–226. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Fu H, Li W, Liu Y, Lao Y, Liu W, Chen C, Yu H, Lee NT, Chang DC, Li P, et al: Mitochondrial proteomic analysis and characterization of the intracellular mechanisms of bis(7)-tacrine in protecting against glutamate-induced excitotoxicity in primary cultured neurons. J Proteome Res. 6:2435–2446. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Jin S, Shen JN, Guo QC, Zhou JG, Wang J, Huang G, Zou CY, Yin JQ, Liu SJ, Liu W, et al: 2-D DIGE and MALDI-TOF-MS analysis of the serum proteome in human osteosarcoma. Proteomics Clin Appl. 1:272–285. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Tang ZX, Chen GX, Liang MY, Rong J, Yao JP, Yang X and Wu ZK: Selective antegrade cerebral perfusion attenuating the TLR4/NF-kB pathway during deep hypothermia circulatory arrest in a pig model. Cardiology. 128:243–250. 2014.PubMed/NCBI

29 

Zrenner B, Ndrepepa G, Karch MR, Schneider MA, Schreieck J, Schömig A and Schmitt C: Electrophysiologic characteristics of paroxysmal and chronic atrial fibrillation in human right atrium. J Am Coll Cardiol. 38:1143–1149. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Huang JL, Lin YJ, Lee PC, Chang HY, Hartono B, Lo LW, Chang SL, Hu YF, Suenari K, Li CH, et al: Right atrial substrate remodeling and atrioventricular node conduction properties in patients with paroxysmal atrial fibrillation. Int J Cardiol. 158:447–449. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Casaclang-Verzosa G, Gersh BJ and Tsang TS: Structural and functional remodeling of the left atrium: Clinical and therapeutic implications for atrial fibrillation. J Am Coll Cardiol. 51:1–11. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Matsuyama TA, Tanaka H, Adachi T, Jiang Y, Ishibashi-Ueda H and Takamatsu T: Intrinsic left atrial histoanatomy as the basis for reentrant excitation causing atrial fibrillation/flutter in rats. Heart Rhythm. 10:1342–1348. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Calò L, Lamberti F, Loricchio ML, De Ruvo E, Colivicchi F, Bianconi L, Pandozi C and Santini M: Left atrial ablation versus biatrial ablation for persistent and permanent atrial fibrillation: A prospective and randomized study. J Am Coll Cardiol. 47:2504–2512. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Kim JB, Bang JH, Jung SH, Choo SJ, Chung CH and Lee JW: Left atrial ablation versus biatrial ablation in the surgical treatment of atrial fibrillation. Ann Thorac Surg. 92:1397–1404; discussion 1404–1405. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Jiang Z, Ma N, Yin H, Ding F, Liu H and Mei J: Biatrial ablation versus limited right atrial ablation for atrial fibrillation associated with atrial septal defect in adults. Surg Today. 45:858–863. 2015. View Article : Google Scholar : PubMed/NCBI

36 

Cooley N, Cowley MJ, Lin RC, Marasco S, Wong C, Kaye DM, Dart AM and Woodcock EA: Influence of atrial fibrillation on microRNA expression profiles in left and right atria from patients with valvular heart disease. Physiol Genomics. 44:211–219. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 14 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
Liu H, Chen G, Zheng H, Qin H, Liang M, Feng K and Wu Z: Differences in atrial fibrillation‑associated proteins between the left and right atrial appendages from patients with rheumatic mitral valve disease: A comparative proteomic analysis Corrigendum in /10.3892/mmr.2018.9218. Mol Med Rep 14: 4232-4242, 2016
APA
Liu, H., Chen, G., Zheng, H., Qin, H., Liang, M., Feng, K., & Wu, Z. (2016). Differences in atrial fibrillation‑associated proteins between the left and right atrial appendages from patients with rheumatic mitral valve disease: A comparative proteomic analysis Corrigendum in /10.3892/mmr.2018.9218. Molecular Medicine Reports, 14, 4232-4242. https://doi.org/10.3892/mmr.2016.5776
MLA
Liu, H., Chen, G., Zheng, H., Qin, H., Liang, M., Feng, K., Wu, Z."Differences in atrial fibrillation‑associated proteins between the left and right atrial appendages from patients with rheumatic mitral valve disease: A comparative proteomic analysis Corrigendum in /10.3892/mmr.2018.9218". Molecular Medicine Reports 14.5 (2016): 4232-4242.
Chicago
Liu, H., Chen, G., Zheng, H., Qin, H., Liang, M., Feng, K., Wu, Z."Differences in atrial fibrillation‑associated proteins between the left and right atrial appendages from patients with rheumatic mitral valve disease: A comparative proteomic analysis Corrigendum in /10.3892/mmr.2018.9218". Molecular Medicine Reports 14, no. 5 (2016): 4232-4242. https://doi.org/10.3892/mmr.2016.5776