Programmed cell death-1 deficiency results in atrial remodeling in C57BL/6 mice

  • Authors:
    • Guoqiang Fu
    • Yizhan Cao
    • Jun Lu
    • Jun Li
    • Li Liu
    • Hongtao Wang
    • Feifei Su
    • Qiangsun Zheng
  • View Affiliations

  • Published online on: December 19, 2012     https://doi.org/10.3892/ijmm.2012.1218
  • Pages: 423-429
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Deficiency of the programmed cell death-1 (PD-1) gene enhances T-cell activation and increases inflammation levels. It has been reported that atrial fibrillation (AF) is closely related to inflammation. The aim of the present study was to investigate the role of PD-1 deficiency in the pathogenesis of AF. Two groups of mice were used in our experiment: the C57BL/6 and the C57BL/6-PD-1-/- group. The expression of the inflammatory cytokines interleukin (IL)-2, -4, -6, -10, -17, interferon-γ and tumor necrosis factor were detected. Furthermore, the levels of atrial myocyte oxidative stress, the atrial effective refractory period (AERP) and the atrial myocardial fibrosis levels were determined. Compared with the C57BL/6 group, we found that the inflammatory cytokines were significantly increased in the PD-1-/- group and the levels of atrial myocyte oxidative stress in the PD-1-/- group were also higher. The AERP became shorter and the dispersion of AERP was increased in the PD-1-/- group. Moreover, the PD-1-/- group presented significant atrial myocardial fibrosis but the C57BL/6 group did not. Our findings strongly suggest that the higher levels of inflammatory cytokines and atrial myocyte oxidative stress were present in the PD-1-/- mice and resulted in atrial electricity and structural remodeling. Due to the atrial remodeling, the PD-1-/- mice were more likely to develop AF.

Introduction

Atrial fibrillation (AF) is the most common arrhythmia in clinical practice, present in 2% of the general population. The prevalence of AF increases markedly with age, affecting as many as 9% of individuals 80 years or older (1). AF has several complications such as thromboembolism and heart failure and is regarded as an independent risk factor for cardiovascular mortality (2).

Numerous studies have focused on the pathogenesis of AF. It is generally accepted that AF may be multifactorial and has been linked to, among others, ectopy from the pulmonary veins, catecholamine release and large atrial size. However, the fundamental arrhythmia mechanisms of AF are rapid ectopic firing and reentrant activity. Atrial remodeling may increase the risk of ectopic or reentrant activity through a multitude of potential mechanisms (3) and is considered to be the new pathophysiological mechanism of AF (4).

In the past few years the relationship between inflammation and AF has drawn significant attention. Inflammation may be a cause of AF and this is supported by the high incidence of AF in post-operative cardiac surgery, a state of severe inflammatory process (57). However, evidence suggests that inflammation may also play a prominent role in nonoperative onset of AF (8).

Programmed cell death-1 (PD-1, Pdcd-1) is a negative immunoreceptor belonging to the CD28/CTLA-4 family. PD-1 deficiency enhances T-cell activation and increases the inflammation level. Previous observations indicated that the PD-1−/− mice with different genetic backgrounds presented different types of autoimmune diseases (9,10). Another study found that PD-1−/− mice cleared adenovirus infections more rapidly but developed more severe hepatocellular injury compared with the control group (11). Meanwhile, the study of Carter et al (12) indicated that the PD-1−/− mice produced a significantly higher concentration of inflammatory cytokines [interferon-γ (IFN-γ), tumor necrosis factor (TNF), interleukin-6 (IL-6) and IL-17]. In the past few years, as an inflammatory animal model, PD-1−/− mice have been used to carry out extensive research, however, none of these studies focused on AF.

In the present study, we compared the expression of inflammatory cytokines, the level of atrial myocyte oxidative stress, the atrial effective refractory period (AERP) and the atrial myocardial fibrosis level of the C57BL/6 PD-1−/− with the C57BL/6 mice. We observed the relationship between the inflammation in the C57BL/6 PD-1−/− mice and the pathogenesis of AF.

Materials and methods

Animals

Two groups of mice were used in our experiment: the C57BL/6 and the C57BL/6-PD-1−/− group (15 male mice, 7–8 weeks old, in each group).

The C57BL/6-PD-1−/− mice were kindly provided by Professor Tasuku Honjo of Kyoto University, Japan. All experiments conformed to the guidelines for the care and use of laboratory animals published by the US National Institutes of Health (NIH Publication no. 85-23, revised 1996). The protocol was approved by the Animal Care and Use Committee of Tangdu Hospital; all animals were maintained under specific pathogen-free conditions prior to the experiment.

Cytometric bead array (CBA) for cytokine assessment

Cytokine assessment was carried out using a mouse Th1/Th2/TH17 cytokine kit (BD Biosciences, San Jose, CA, USA) for simultaneous detection of seven cytokines (IL-2, -4, -6, -10, -17A, TNF and IFN-γ). The kit performance was optimized for analysis of physiologically relevant concentrations (pg/ml level) of specific cytokine proteins in serum samples.

The CBA technique utilizes micro particles or beads dyed with discrete fluorescence intensity. The dye incorporated in the beads fluoresces strongly at 650 nm (measured as FL4 signals in the BD FACSCalibur flow cytometer) when excited with an argon laser. Each particle population of a given intensity represents a discrete population for constructing an immunoassay for a single analyte and each particle is covalently coupled with an antibody directed against a specific analyte. When these capture beads for different analytes are used as a mixture, the level of individual analyte within samples can be measured simultaneously. Detection is mediated by the binding of specific detection antibodies that are directly conjugated with phycoerythrin (PE), thus providing an FL2 fluorescent signal on the appropriate bead. This signal is proportional to the concentration of the analyte (Fig. 1).

Mouse serum (50 μl) and PE detection antibody were incubated with capture bead reagent for 2 h in the dark at room temperature. All unbound antibodies were washed (1.0 ml wash buffer), re-suspended in 250 μl before acquiring data on the BD FACSCalibur bio-analyzer (BD Biosciences).

By dedicated CBA analysis software, seven individual cytokine standard curves were used to determine the concentration of each analyte in the test sample; detection was calculated from curve estimation for an average of ten assays using power fit and R2>0.99 for all cytokines (Fig. 2).

Cell isolation

The isolation of single atrial myocyte from mice was performed as described by Cho et al (13). Mice were sacrificed by cervical dislocation, and the heart was quickly removed. The heart was cannulated by a 24-gauge needle and then retrogradely perfused via the aorta on a Langendorff apparatus. During coronary perfusion, all perfusates were maintained at 37°C and equilibrated with 100% O2. Initially the heart was perfused with normal Tyrode solution for 2–3 min to clear the blood. The heart was then perfused with Ca2+-free solution for 2 min. Finally, the heart was perfused with enzyme solution for 15 min. The enzyme solution contained 0.15 mg/ml collagenase (Sigma-Aldrich) in Ca2+-free solution. Following perfusion with enzyme solution, the atria were separated from the ventricles and chopped into small pieces. Single cells were dissociated in storage medium from these small pieces using a blunt-tip glass pipette.

Generation of reactive oxygen species (ROS)

Generation of myocardial cell intracellular ROS was measured using the fluorescent dye 2′,7′-dichlorofluorescein diacetate (DCFH-DA) (Sigma-Aldrich). DCFH-DA is a non-polar and non-fluorescent compound that can diffuse into the cell where it is deacetylated by cellular esterases into a non-fluorescent polar derivative DCFH that is impermeable to the cell membrane. DCFH is rapidly oxidized to the highly fluorescent dichlorofluorescein (DCF) in the presence of intracellular ROS and can be analyzed with excitation 495 nm/emission 525 nm (measured in the FL1 channel).

Atrial myocytes were seeded in 96-well black tissue culture microplates. The cells received DCFH-DA (2 μmol/l) for 20 min at 37°C. Following removal of the DCFH-DA, the microplate was incubated at 37°C for 1 h (14), and data was acquired on a BD FACSCalibur bioanalyzer (BD Biosciences).

Atrial effective refractory periods (AERPs)

The electrophysiology research was performed according to Etzion et al (15). All mice were anesthetized with intraperitoneal injection of sodium pentobarbital (50 mg/kg). Following intubation and mechanical ventilation, the chest was opened through the right fourth intercostal space, and two bipolar screws-in pacing lead were fixed in proper sequence to four sites in the right atrial as follows: appendage, high lateral, low lateral and anterior walls. During ventilation, the arterial blood gases were adjusted to between pH 7.35 and 7.45.

Electrograms were recorded in bipolar mode at a filter setting of 30–500 Hz and stored digitally on a custom acquisition system (Quinton Electrophysiology, Canada). A programmed stimulator (Fukuda Denshi BC02A, Japan) was used to deliver square-wave impulses of 1-ms duration. The AERPs were measured at each of two sites at basic cycle lengths (100 ms) after 30 sec of continuous pacing to achieve a steady state; an atrial extra-stimulus was introduced after every six drive beat, and all stimuli were twice the diastolic threshold. The initial extra-stimulus coupling interval was set at 30 ms, and the coupling interval was set in steps of 5 ms increasing after every six beat until the extra-stimulus resulted in atrial capture. The coupling interval was then reduced by 5 ms and increased in steps of 1 ms until atria captured again by extra-stimulus. AERP was defined as the longest S1S2 coupling interval that failed to lead to atrial capture (Fig. 3). Dispersion of AERP (dAERP) equaled to maximum AERP minus minimum AERP among the four sides.

Histological examination

Atrial cardiac muscle was dehydrated by storing in 70% ethanol, cleared and embedded in paraffin. Routine 4 μm serial section was performed, and slides were dried in a 60°C oven. For Masson staining, slides were submerged in Masson trichrome solution (the Fourth Military Medical University Pathology Laboratory, Xi’an, China) for 5 min, washed with 0.2% acetic acid for 10 sec, followed by 5% phosphotungstic acid for 10 min, then washed twice with 0.2% acetic acid solution, stained by 2% aniline blue solution for 5 min, washed twice by 0.2% acetic acid, dehydrated using gradient ethanol, cleared in xylene and sealed using neutral Balsam.

Statistical analysis

All statistical analyses were performed using SPSS software (version 18; SPSS Inc., Chicago, IL, USA). Data are expressed as the means ± SD and compared by the two-sample Student’s t-test. Differences were considered to be statistically significant when P<0.05.

Results

Inflammatory cytokines

There were significant differences in inflammatory cytokine levels between the two groups. The inflammatory cytokine levels of PD-1−/− mice were significantly higher than those of the C57BL/6 mice. IL-10, 1212.35±300.42 vs. 208.8±57.25 pg/ml, P<0.001 (Fig. 4A); IL-6, 165.21±42.49 vs. 35.47±6.04 pg/ml, P<0.001 (Fig. 4B); IL-2, 3.95±1.06 vs. 2.8±0.63 pg/ml, P<0.01; IL-4, 7.19±1.07 vs. 3.11±0.35 pg/ml, P<0.001; IFN-γ, 22.1±4.72 vs. 5.17±1.27 pg/ml, P<0.001; TNF, 36.49±8.21 vs. 24.55±4.29 pg/ml, P<0.001; IL-17A, 42.03±8.53 vs. 8.05±1.76 pg/ml, P<0.001 (Fig. 4C).

Oxidative stress

Intracellular ROS levels were measured by fluorescent probe DCFH-DA (Fig. 5). There were significant differences in the mean DCF fluorescence intensity between two groups (30.21±1.66 vs. 24.35±1.82, P<0.001).

AERPs

AERPs at each site were significantly shortened in PD-1−/− mice compared with the C57BL/6 mice (Table I). We also found that the dAERP in PD-1−/− mice was significantly increased, compared with the C57BL/6 mice, 2.73±1.44 vs. 1.53±0.92 ms, P<0.05.

Table I.

Comparison of AERP between the C57BL/6 and the PD-1 deficient mice.

Table I.

Comparison of AERP between the C57BL/6 and the PD-1 deficient mice.

AERPs BCL (ms)C57BL/6 (n=15)PD-1 deficient (n=15)
RAA 10042.47±0.7441.73±1.03a
LRA 10042.87±0.9941.47±2.17a
HRA 10042.80±0.9441.87±1.19a
ARA 10042.60±1.4541.53±1.30a

{ label (or @symbol) needed for fn[@id='tfn1-ijmm-31-02-0423'] } AERP, atrial effective refractory period; RAA, right atrial appendage; LRA, low lateral right atrium; HRA, high lateral right atrium; ARA, anterior right atrium.

a P<0.05 compared with the control group.

Pathological examination

Histological studies were performed to verify the potential pathological substrate difference between the two groups. Atrial myocardial fibrosis was detected in the PD-1−/− group, but not in the C57BL/6 group (Fig. 6).

Discussion

PD-1−/− mice and cardiovascular diseases

Programmed cell death-1 (PD-1) is an inhibitory receptor in the CD28/CTLA-4 family and it can be inducibly expressed on CD8 and CD4 T-cells, natural killer T-cells, B-cells and activated monocytes (9,10,16). The ligands for PD-1 (PD-Ls) are PD-L1 and PD-L2. PD-Ls pathways control the induction and maintenance of peripheral T-cell tolerance (10). PD-1 deficiency enhances T-cell activation and increases the inflammation level.

PD-1−/− mice were first reported by Nishimura et al (17) in 1998. Experiments showed that the PD-1−/− mice with different genetic backgrounds presented different types of diseases and several were cardiovascular diseases. Nishimura et al (18) found that some 2C-PD-1−/−H-2b/d mice died of a Graft-Versus-Host-like (GVH-like) disease; the survivor mouse myocardium presented inflammatory cell infiltration. Studies also indicated that the heart of BALB/c-PD-1−/− mice showed various degrees of inflammation with marked deposition of immune complex on the surface of cardiomyocytes, and their sera contained high titer auto-antibodies against cardiac troponin I (cTnI) (1921). In addition, MRL-PD-1−/− mice developed fatal myocarditis (22), massive infiltration of CD4+ and CD8+ T-cells and myeloid cells were found in their hearts concomitant with the production of high-titer auto-antibodies against cardiac myosin (22).

These studies indicated that the PD-1−/− mice are closely associated with cardiovascular diseases; the basic mechanism is the high system inflammation level resulting in the inflammatory cell infiltration and/or the generation of auto-antibodies against cardiac tissue.

In the present study, we found that the PD-1−/− group presented atrial electricity remodeling (shorter AERP and increased dAERP) and structural remodeling (atrial myocardial fibrosis). It is generally accepted that AF results from the presence of multiple reentrant circuits originating in the atria (23), and atrial remodeling increases the probability of generating multiple atrial wavelets by dispersion of refractoriness and rapid atrial activation (4). Our findings strongly suggest that the PD-1−/− mice are more likely to develop AF.

Inflammatory cytokines, reactive oxygen species (ROS) and atrial remodeling

Previous studies indicated that inflammation may lead to ‘atrial myocarditis’; atrial electrical and structural remodeling resulting from it subsequently led to the initiation and maintenance of AF (24,25).

Other studies indicated that T lymphocytes participated in the cardiac remodeling (26,27) and Th1 and Th2 responses are involved in the most fiber proliferative disorders. Th1 cytokines (IFN-γ, TNF and IL-2) are considered to be involved in the initiation phase of fibrosis and the Th2 cytokines (IL-4, -6 and -10) during the latter stages (28). Th17 cells were characterized by the production of IL-17 as signature cytokines and also participate in the process of cardiac remodeling. Studies have shown that IL-17 could promote the production of collagen in cardiac fibroblasts (29,30).

Inflammation is also involved in the process of electrical remodeling. Atrial inflammation following cardiac surgery could influence the electrical properties of atrial tissue, and the level of atrial inflammation was associated with a proportional increase in the inhomogeneity of atrial conduction and AF duration (31). In this process, inflammation cytokines may play an important role. Previous studies found that overexpression of TNF could downregulate connexin 40 (32), Kv4.2, Kv4.3 and Kv1.5 and KChIP-2 (33,34). Downregulation of connexin 40 slows down the conduction and increases the susceptibility of atrial arrhythmias, decreases potassium channel-interacting protein, reduces Ito (transient outward K current), IK, slow1, and IK, slow2, which attenuate the effective refractory period (ERP).

In the present study, although two groups of mice lived in the same environment and were fed the same food and water, the pathogen existing in the environment including microbe, parasites and virus aggravated the inflammation process in PD-1−/− mice. Therefore, the PD-1−/− group presented higher levels of inflammatory cytokines.

Oxidative stress refers to the total burden of potentially harmful ROS formed during cellular metabolism. High oxidative stress levels may be closely related to atrial electrical remodeling. Carnes et al (35) found that atrial tachy-pacing provoked increased protein nitration indicating enhanced oxidative stress in a dog model and the process was accompanied by a decrease in the ERP. It also showed that pretreatment of the animals with oral vitamin C (a water-soluble antioxidant) attenuated the ERP shortening (36). In addition, another study indicated that ROS also played an important role in cardiac remodeling by angiotensin II (37).

In several pathophysiological conditions, inflammation can augment oxidative stress and vice versa. Therefore, some people speculate that inflammation and oxidative stress cooperate at some level facilitating atrial remodeling (25,38).

In the present study, we found that the PD-1−/− group presented higher levels of inflammatory cytokines compared with the C57BL/6 group. At the same time, intracellular ROS of cardiac atrial myocytes were also higher. They may be interrelated and result in shorter AERPs, increased dAERP and myocardial fibrosis.

Our study is the first to prove that PD-1 deficiency results in atrial electrical and structural remodeling, raising the vulnerability of C57BL/6 mice to AF. However, it should be noted that this was an animal study with a special gene background and should not be over-interpreted. A clinical trial is required to confirm that the systemic inflammation causes the onset of AF in nonoperative settings. Sata et al (39) observed 15 patients with paroxysmal AF and 11 patients with normal sinus rhythm. CRP, IL-6 and TNF-α were measured at baseline, 24 h, and two weeks after cardioversion, and they found that CRP, IL-6 and TNF-α were greater in the AF group and did not normalize two weeks after cardioversion. The study suggested that inflammation may be an independent risk factor for AF, however their sample size was limited.

Tang et al (40) hypothesized that Chlamydia pneumoniae infection was a possible cause of AF by initiating inflammation response. They found that there was a close epidemiological and pathological relationship between both sides, but the hypothesis remains to be confirmed by clinical experiments. Bigger sample clinical trials need to be performed to validate the role of systemic inflammation in the onset of nonoperative AF.

In conclusion, PD-1 deficiency resulted in a significant increase in inflammatory cytokines and intracellular ROS levels in C57BL/6 mice. Higher inflammatory cytokines and intracellular ROS levels led to atrial remodeling, and, due to atrial remodeling, the C57BL/6 PD-1−/− mice may be more susceptible to AF.

Acknowledgements

The authors thank Professor Tasuku Honjo for kindly providing us with the C57BL/6 PD-1−/− mice. This study was supported by the National Natural Science Foundation of China (no. 81070161).

References

1. 

Page RL: Clinical practice. Newly diagnosed atrial fibrillation. N Engl J Med. 351:2408–2416. 2004. View Article : Google Scholar : PubMed/NCBI

2. 

Benjamin EJ, Wolf PA, D’Agostino RB, Silbershatz H, Kannel WB and Levy D: Impact of atrial fibrillation on the risk of death: the Framingham Heart Study. Circulation. 946–952. 1998. View Article : Google Scholar : PubMed/NCBI

3. 

Nattel S, Burstein B and Dobrev D: Atrial remodeling and atrial fibrillation: mechanisms and implications. Circ Arrhythm Electrophysiol. 1:62–73. 2008. View Article : Google Scholar : PubMed/NCBI

4. 

Sharma D, Li G, Xu G, Liu Y and Xu Y: Atrial remodeling in atrial fibrillation and some related microRNAs. Cardiology. 120:111–121. 2011. View Article : Google Scholar : PubMed/NCBI

5. 

Bruins P, te Velthuis H, Yazdanbakhsh AP, et al: Activation of the complement system during and after cardiopulmonary bypass surgery: postsurgery activation involves C-reactive protein and is associated with postoperative arrhythmia. Circulation. 96:3542–3548. 1997. View Article : Google Scholar

6. 

Gabay C and Kushner I: Acute-phase proteins and other systemic responses to inflammation. N Engl J Med. 340:448–454. 1999. View Article : Google Scholar : PubMed/NCBI

7. 

Ommen SR, Odell JA and Stanton MS: Atrial arrhythmias after cardiothoracic surgery. N Engl J Med. 336:1429–1434. 1997. View Article : Google Scholar : PubMed/NCBI

8. 

Engelmann MD and Svendsen JH: Inflammation in the genesis and perpetuation of atrial fibrillation. Eur Heart J. 26:2083–2092. 2005. View Article : Google Scholar : PubMed/NCBI

9. 

Greenwald RJ, Freeman GJ and Sharpe AH: The B7 family revisited. Annu Rev Immunol. 23:515–548. 2005. View Article : Google Scholar : PubMed/NCBI

10. 

Okazaki T and Honjo T: The PD-1-PD-L pathway in immunological tolerance. Trends Immunol. 27:195–201. 2006. View Article : Google Scholar : PubMed/NCBI

11. 

Iwai Y, Terawaki S, Ikegawa M, Okazaki T and Honjo T: PD-1 inhibits antiviral immunity at the effector phase in the liver. J Exp Med. 198:39–50. 2003. View Article : Google Scholar : PubMed/NCBI

12. 

Carter LL, Leach MW, Azoitei ML, et al: PD-1/PD-L1, but not PD-1/PD-L2, interactions regulate the severity of experimental autoimmune encephalomyelitis. J Neuroimmunol. 182:124–134. 2007. View Article : Google Scholar : PubMed/NCBI

13. 

Cho H, Nam GB, Lee SH, Earm YE and Ho WK: Phosphatidylinositol 4,5-bisphosphate is acting as a signal molecule in alpha(1)-adrenergic pathway via the modulation of acetylcholine-activated K(+) channels in mouse atrial myocytes. J Biol Chem. 276:159–164. 2001.PubMed/NCBI

14. 

Golan O, Issan Y, Isak A, Leipziger J, Robaye B and Shainberg A: Extracellular nucleotide derivatives protect cardiomyocytes against hypoxic stress. Biochem Pharmacol. 81:1219–1227. 2011. View Article : Google Scholar : PubMed/NCBI

15. 

Etzion Y, Mor M, Shalev A, et al: New insights into the atrial electrophysiology of rodents using a novel modality: the miniature-bipolar hook electrode. Am J Physiol Heart Circ Physiol. 295:H1460–H1469. 2008. View Article : Google Scholar : PubMed/NCBI

16. 

Chen L: Co-inhibitory molecules of the B7-CD28 family in the control of T-cell immunity. Nat Rev Immunol. 4:336–347. 2004. View Article : Google Scholar : PubMed/NCBI

17. 

Nishimura H, Minato N, Nakano T and Honjo T: Immunological studies on PD-1 deficient mice: implication of PD-1 as a negative regulator for B cell responses. Int Immunol. 10:1563–1572. 1998. View Article : Google Scholar : PubMed/NCBI

18. 

Nishimura H, Nose M, Hiai H, Minato N and Honjo T: Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 11:141–151. 1999. View Article : Google Scholar : PubMed/NCBI

19. 

Nishimura H, Okazaki T, Tanaka Y, et al: Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 291:319–322. 2001. View Article : Google Scholar : PubMed/NCBI

20. 

Okazaki T and Honjo T: Pathogenic roles of cardiac autoantibodies in dilated cardiomyopathy. Trends Mol Med. 11:322–326. 2005. View Article : Google Scholar : PubMed/NCBI

21. 

Okazaki T, Tanaka Y, Nishio R, et al: Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med. 9:1477–1483. 2003. View Article : Google Scholar : PubMed/NCBI

22. 

Wang J, Okazaki IM, Yoshida T, et al: PD-1 deficiency results in the development of fatal myocarditis in MRL mice. Int Immunol. 22:443–452. 2010. View Article : Google Scholar : PubMed/NCBI

23. 

Falk RH: Atrial fibrillation. N Engl J Med. 344:1067–1078. 2001. View Article : Google Scholar : PubMed/NCBI

24. 

Aviles RJ, Martin DO, Apperson-Hansen C, et al: Inflammation as a risk factor for atrial fibrillation. Circulation. 108:3006–3010. 2003. View Article : Google Scholar : PubMed/NCBI

25. 

Korantzopoulos P, Kolettis T, Siogas K and Goudevenos J: Atrial fibrillation and electrical remodeling: the potential role of inflammation and oxidative stress. Med Sci Monit. 9:225–229. 2003.PubMed/NCBI

26. 

Jakubzick C, Choi ES, Kunkel SL, Joshi BH, Puri RK and Hogaboam CM: Impact of interleukin-13 responsiveness on the synthetic and proliferative properties of Th1- and Th2-type pulmonary granuloma fibroblasts. Am J Pathol. 162:1475–1486. 2003. View Article : Google Scholar : PubMed/NCBI

27. 

Liu W, Feng W, Wang F, et al: Adenovirus-mediated ICOSIg gene transfer alleviates cardiac remodeling in experimental autoimmune myocarditis. Immunol Cell Biol. 86:659–665. 2008. View Article : Google Scholar : PubMed/NCBI

28. 

Yu Q, Horak K and Larson DF: Role of T lymphocytes in hypertension-induced cardiac extracellular matrix remodeling. Hypertension. 48:98–104. 2006. View Article : Google Scholar : PubMed/NCBI

29. 

Cortez DM, Feldman MD, Mummidi S, et al: IL-17 stimulates MMP-1 expression in primary human cardiac fibroblasts via p38 MAPK- and ERK1/2-dependent C/EBP-beta, NF-kappaB, and AP-1 activation. Am J Physiol Heart Circ Physiol. 293:H3356–H3365. 2007. View Article : Google Scholar : PubMed/NCBI

30. 

Feng W, Li W, Liu W, Wang F, Li Y and Yan W: IL-17 induces myocardial fibrosis and enhances RANKL/OPG and MMP/TIMP signaling in isoproterenol-induced heart failure. Exp Mol Pathol. 87:212–218. 2009. View Article : Google Scholar : PubMed/NCBI

31. 

Ishii Y, Schuessler RB, Gaynor SL, et al: Inflammation of atrium after cardiac surgery is associated with inhomogeneity of atrial conduction and atrial fibrillation. Circulation. 111:2881–2888. 2005. View Article : Google Scholar : PubMed/NCBI

32. 

Sawaya SE, Rajawat YS, Rami TG, et al: Downregulation of connexin40 and increased prevalence of atrial arrhythmias in transgenic mice with cardiac-restricted overexpression of tumor necrosis factor. Am J Physiol Heart Circ Physiol. 292:1561–1567. 2007. View Article : Google Scholar : PubMed/NCBI

33. 

Kawada H, Niwano S, Niwano H, et al: Tumor necrosis factor-alpha downregulates the voltage gated outward K+ current in cultured neonatal rat cardiomyocytes: a possible cause of electrical remodeling in diseased hearts. Circ J. 70:605–609. 2006. View Article : Google Scholar : PubMed/NCBI

34. 

Petkova-Kirova PS, Gursoy E, Mehdi H, McTiernan CF, London B and Salama G: Electrical remodeling of cardiac myocytes from mice with heart failure due to the overexpression of tumor necrosis factor-alpha. Am J Physiol Heart Circ Physiol. 290:2098–2107. 2006. View Article : Google Scholar : PubMed/NCBI

35. 

Carnes CA, Chung MK, Nakayama T, et al: Ascorbate attenuates atrial pacing-induced peroxynitrite formation and electrical remodeling and decreases the incidence of postoperative atrial fibrillation. Circ Res. 89:e32–e38. 2001. View Article : Google Scholar

36. 

Galaris D and Korantzopoulos P: On the molecular mechanism of metmyoglobin-catalyzed reduction of hydrogen peroxide by ascorbate. Free Radic Biol Med. 22:657–667. 1997. View Article : Google Scholar : PubMed/NCBI

37. 

Wu S, Gao J, Ohlemeyer C, et al: Activation of AP-1 through reactive oxygen species by angiotensin II in rat cardiomyocytes. Free Radic Biol Med. 39:1601–1610. 2005. View Article : Google Scholar : PubMed/NCBI

38. 

Korantzopoulos P, Galaris D, Papaioannides D and Kokkoris S: C-reactive protein and oxidative stress in atrial fibrillation. Int J Cardiol. 88:103–104. 2003. View Article : Google Scholar : PubMed/NCBI

39. 

Sata N, Hamada N, Horinouchi T, et al: C-reactive protein and atrial fibrillation. Is inflammation a consequence or a cause of atrial fibrillation? Jpn Heart J. 45:441–445. 2004. View Article : Google Scholar : PubMed/NCBI

40. 

Tang RB, Dong JZ, Liu XP and Ma CS: Inflammation and atrial fibrillation: is Chlamydia pneumoniae a candidate pathogen of atrial fibrillation. Med Hypotheses. 67:462–466. 2006.PubMed/NCBI

Related Articles

Journal Cover

February 2013
Volume 31 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Fu G, Cao Y, Lu J, Li J, Liu L, Wang H, Su F and Zheng Q: Programmed cell death-1 deficiency results in atrial remodeling in C57BL/6 mice. Int J Mol Med 31: 423-429, 2013
APA
Fu, G., Cao, Y., Lu, J., Li, J., Liu, L., Wang, H. ... Zheng, Q. (2013). Programmed cell death-1 deficiency results in atrial remodeling in C57BL/6 mice. International Journal of Molecular Medicine, 31, 423-429. https://doi.org/10.3892/ijmm.2012.1218
MLA
Fu, G., Cao, Y., Lu, J., Li, J., Liu, L., Wang, H., Su, F., Zheng, Q."Programmed cell death-1 deficiency results in atrial remodeling in C57BL/6 mice". International Journal of Molecular Medicine 31.2 (2013): 423-429.
Chicago
Fu, G., Cao, Y., Lu, J., Li, J., Liu, L., Wang, H., Su, F., Zheng, Q."Programmed cell death-1 deficiency results in atrial remodeling in C57BL/6 mice". International Journal of Molecular Medicine 31, no. 2 (2013): 423-429. https://doi.org/10.3892/ijmm.2012.1218