Abstract
Objective:
Primary mitral regurgitation is a valvular lesion in which the left ventricular ejection fraction remains preserved for long periods, delaying a clinical trigger for mitral valve intervention. In this study, we sought to investigate whether adverse left ventricular remodeling occurs before a significant fall in ejection fraction and characterize these changes.
Methods:
Sixty-five rats were induced with severe mitral regurgitation by puncturing the mitral valve leaflet with a 23-G needle using ultrasound guidance. Rats underwent longitudinal cardiac echocardiography at biweekly intervals and hearts explanted at 2 weeks (n = 15), 10 weeks (n = 15), 20 weeks (n = 15), and 40 weeks (n = 15). Sixty age- and weight-matched healthy rats were used as controls. Unbiased RNA-sequencing was performed at each terminal point.
Results:
Regurgitant fraction was 40.99 9.40%, with pulmonary flow reversal in the experimental group, and none in the control group. Significant fall in ejection fraction occurred at 14 weeks after mitral regurgitation induction. However, before 14 weeks, end-diastolic volume increased by 93.69 52.38% (P < .0001 compared with baseline), end-systolic volume increased by 118.33 47.54% (P < .0001 compared with baseline), and several load-independent pump function indices were reduced. Transcriptomic data at 2 and 10 weeks before fall in ejection fraction indicated up-regulation of myocyte remodeling and oxidative stress pathways, whereas those at 20 and 40 weeks indicated extracellular matrix remodeling.
Conclusions:
In this rodent model of mitral regurgitation, left ventricular ejection fraction was preserved for a long duration, yet rapid and severe left ventricular dilatation, and biological remodeling occurred before a clinically significant fall in ejection fraction.
Keywords: mitral valve prolapse, mitral regurgitation, ventricular remodeling, primary mitral regurgitation, neo-chordoplasty, ejection fraction, heart murmur
Graphical Abstract
Rapid LV remodeling occurs despite preserved ejection fraction in mitral regurgitation.
Primary mitral regurgitation (MR) resulting from myxomatous degenerative valve disease occurs in 1.7% of the American populace and is the second most common form of valvular heart disease.1–3 The regurgitation from the highpressure left ventricle into the low impedance left atrium reduces forward stroke volume, elevates left ventricular (LV) filling pressures, and imposes a chronic, low-pressure, volume overload on the LV.4 The LVadapts to this hemodynamic stress with acute hypercontractility but progressive eccentric remodeling, manifesting primarily as LV dilatation and increased sphericity. Ejection fraction(EF) is largely preserved for prolonged periods, and thus referral to mitral valve diagnosed,5,6 Although this strategy is currently recommended, patients who undergo mitral valve repair or replacement present with paradoxical new-onset LV dysfunction after correction of MR.7 This could indicate that the functional deficit in the LV before the mitral valve surgery is masked by the low impedance flow into the left atrium, keeping the EF high despite poor function. As the risk associated with mitral valve repair falls, a push toward earlier intervention before a fall in EF is proposed.8 However, identifying that earlier time point would require an understanding of the natural history of remodeling after the onset of MR, which is currently lacking. In this study, we sought to develop a animal model of severe MR and test the hypothesis that significant LV chamber dilatation and reduction in load independent contractile indices occurs before a significant fall in EF. Toward this end, in this study, we used a controlled rodent model of MR9,10 to investigate hereto unknown longitudinal changes in LV chamber volume and shape, contractile function, and the myocardial transcriptome at 2, 10, 20, and 40 weeks (Figure 1).
FIGURE 1.
Top row, Schematic depicting the study design and sample sizes used in each group. Bottom row, Salient findings from the study describing the effect of mitral regurgitation on the function, structure and transcriptome of the left ventricular myocardium.
METHODS
Ethical Statement
Use of animals was approved by the Institutional Animal Care and Use Committee at Emory University and performed per the guidelines of the National Institutes of Health. Animals were purchased from a single vendor (Envigo, Indianapolis, Ind), housed in a temperature- and humidity-controlled environment with a 12:12-hour light cycle, at an Association for Assessment and Accreditation of Laboratory Animal Care–accredited facility.
Animal Selection
Adult, male, Sprague–Dawley rats (n = 125, 350–400 g) were used and socially housed prior to surgery and singly housed after surgery, with continuous access to drinking water and rat chow. Rats were randomized to be induced with severe MR (n = 65) or act as sham controls (n = 60). Sample sizes were estimated to test the hypothesis that severe MR will cause a 25% greater end-diastolic volume (EDV) compared with sham rats at each time point, with 80% power and an alpha of 0.05.
Surgical Procedures
Procedures to induce severe MR were described earlier.10,11 Rats were sedated, intubated, and mechanically ventilated. Two percent to 2.5% isoflurane mixed in 100% oxygen was used for anesthesia, body temperature was maintained at 37°C, and an electrocardiogram was acquired (Mouse Monitor; Indus Instruments, Webster, Tex). Carprofen (2.5 mg/kg, subcutaneously [SQ]), gentamicin (6 mg/kg, SQ), and sterile saline (1 mL, SQ) were then administered. With the rat in a right decubitus position, a left thoracotomy was performed in the fifth intercostal space to access the heart. The apex was stabilized with a purse-string suture (6–0 PROLENE), and an 8-Fr intracardiac echo probe was inserted into the esophagus to achieve a 2-chamber view of the heart (Figure 2, A1). A 23-G needle was inserted through the apical purse-string into the anterior mitral leaflet (Figure 2, A2–3), creating a hole and severe MR (Figure 2, B1). The purse-string was gently tightened, the thoracotomy was closed, and a temporary chest tube was used. Rats were extubated, carprofen (2.5 mg/kg, SQ) was immediately administered, and Buprenex (0.02 mg/kg, SQ) was administered within 3 hours. Postoperative pain was managed with carprofen (SQ, 5 mg/kg, postoperative days 1–3) and gentamicin (SQ, 6 mg/kg, postoperative days 1–3), and Buprenex (SQ, 0.05 mg/kg, as needed). At 2 weeks after the surgery, severity of MR was assessed by transesophageal echocardiography (TEE; Figure 2, B1–3), and the rats were followed to 2, 10, 20, or 40 weeks (n = 15 per group). In human years, these time points scale to 1, 5, 10, and 20 human years.12 In the sham group, 15 age- and weight-matched rats that underwent surgery were followed and terminated at the same time points. The entire procedure is depicted in Video 1.
FIGURE 2.
Rodent model of severe MR using an image-guided needle technique to puncture the mitral valve leaflet. A1, The animal is placed in right decubitus position, with an intracardiac ultrasound probe inserted into the esophagus to enable transesophageal imaging. A2, Ultrasound image depicting a needle inserted into the beating heart via the ventricular apex. A3,With ultrasound guidance, the needle tip is advanced into the mitral valve leaflet to perforate it. B1-B3, Induction of MR is validated in real time using color Doppler imaging of the regurgitant jet, pulsed-wave Doppler of flow reversal, and pulmonary vein flow reversal. B4, In a representative rat heart explanted at the end of the experiment, the perforation in the anterior leaflet is depicted. The hole is defined and matches the size of the needle. C1, Severe MR was confirmed in all the ratsat 2 weeks after thesurgery. The severity of regurgitation is quantified hereby measuring the areaofthe MRjet and normalizing it to the left atrial area. C2, Regurgitant volume was also calculated using the Doppler data obtained from the rodents and using the area of the regurgitant orifice, confirming with another technique that the mitral regurgitation is severe. C3, An indirect approach to confirming the severity of regurgitation is depicted, wherein reversal of the pulmonary vein flow due to acute elevation of the left atrial pressure from regurgitation is noted. The S/D ratio is positive in the sham animals, whereas it is reversed in the rats with mitral regurgitation. C4, Survivalcurves are presented for the rats that completed 40weeks of follow-up in both the MR and sham groups. The color-shaded area represents the 95% confidence limits. MR, Mitral regurgitation; S/D, systolic-diastolic.
Cardiac Echocardiography
Transthoracic echocardiography and TEE were performed just before surgery (baseline) and at biweekly intervals after surgery, until termination. Transthoracic echocardiography was performed with the rats under mild sedation, and parasternal long-axis and short-axis B-mode and M-mode images were obtained with a 21-MHz rat probe on a Visualsonics 2100 system (FIJUFILM Visualsonics, Inc, Tokyo, Japan). Endocardial and epicardial borders were traced to calculate LV volumes, EF, LV mass, wall thickness, and fractional wall thickening by 2 blinded users and interobserver variability is quantified (Tables E1 and E2). TEE was performed with the rats intubated and anesthetized, and using an 8-Fr intracardiac echo probe (8 MHz, Acunav; Biosense Webster, Inc, Irvine, Calif). High esophageal views of the chambers and mitral valve and color Doppler images were obtained. Pulsed-wave Doppler imaging was performed at the mitral valve and the pulmonary vein to assess flows. MR severity was evaluated using 3 quantification methods: MR jet area (%), regurgitant volume, and systolic pulmonary systolic-diastolic ratio. MR jet area (%) was measured by tracing the regurgitant jet on color Doppler and normalized to left atrial area. Severe MR jet area (%) was defined as ≥30%, as per the guidelines of the American Society of Echocardiography.13 Regurgitant volume was calculated by multiplying the MR velocity time integral measured on pulsed wave Doppler by the area of the circular regurgitant orifice created by the 23-G needle (0.64 mm outer diameter). Regurgitant volume 80 mL was considered severe MR. Pulmonary flow reversal was assessed as the ratio of the systolic (Swave) and diastolic (D-wave) velocities. Severe MR was defined by a systolic blunting or total reversal of the systolic-diastolic wave ratio.
Invasive Hemodynamic Measurements
Pressure volume (PV) loops were obtained in all rats at termination, with a 1.9-Fr transapical admittance catheter (ADV500; Transonic Systems, London, Ontario, Canada) under 10 to 15 seconds of apnea. Preload reduction was achieved by compressing the inferior vena cava. End-systolic and end-diastolic pressure volume relationships and other pump function indices, such as EDV versus stroke work (preload recruitable stroke work), maximum dP/dt versus EDV, and pressure volume loop area (PVA) versus EDV were computed.
Tissue Collection and RNA Seq
Rats were heparinized and terminated under deep anesthesia, with 100 mg/kg Euthasol (390 mg/mL pentobarbital sodium mixed with 50 mg/mL phenytoin sodium). The heart and lungs were immediately excised, photographed, and weighed. The LV free wall was separated, and the mid-ventricular region was snap frozen. Total RNA was extracted from 5 samples in each group using the miRNeasy Mini Kit (Yerkes NHP Genomics Core, Emory University, Atlanta, Ga). Postquality check, RNA-sequencing was performed using the Illumina platform to generate unaligned reads (Novogene, Sacramento, Calif). RNAseq data analysis was performed using Partek Flow (Partek Inc, St Louis, Mo; build 4.0.15). Version (rn6) rat reference genome were used to map reads. The post-alignment QC module of Partek Flow was used to visualize the average base quality score per position as well as the mapping quality per alignment. The mapped reads were quantified using the RefSeq transcripts-2019–11-01 annotation for quantification using the Partek E/M method to determine the differential gene expression using gene-specific analysis comparing each experimental group against sham. Significant differentially expressed genes were shortlisted using a cutoff of P <.05 and 2 ≤ fold change ≤ −2. Functional enrichment analysis was performed using Protein Analysis Through Evolutionary Relationships (version 15), Gene Ontology, and Kyoto Encyclopedia of Genes and Genomes databases, to identify molecular and biochemical pathways. All RNA-seq data have been deposited in the NCBI Gene Expression Omnibus database with experiment series accession number GSE148559.
Statistical Analysis
Prism 7.0 (GraphPad Software, Inc, La Jolla, Calif) was used for data analysis. Data following a gaussian distribution are depicted as mean 1 standard deviation, and non-normal data are represented as median with interquartile range. MR severity was compared using a paired t test for normally distributed data, and a Wilcoxon matched-pairs test for nonparametric data, comparing baseline and 2-week values. To compare longitudinal echocardiography data (EDV, end-systolic volume [ESV], stroke volume [SV], EF) from the sham and MR group, a repeated-measure 2-way analysis of variance with Tukey’s multiple comparisons was performed, when all data points were available. For all other echo and PV loop data, a one-way analysis of variance with Tukey’s multiple comparisons test was used to compare groups with normally distributed data. Mann-Whitney U test with Dunn’s correction and multiple comparisons was used for nonparametric data.
RESULTS
Validation of Severe MR and Overall Mortality
There were no procedural complications or mortality associated with the MR surgery. Two weeks after surgery, MR jet area normalized to left atrial area was 40.99 ± 9.40%, compared with 0.25 ± 1.34% at baseline (P < .0001) (Figure 2, C1). Mitral regurgitant volume was 119.50 ± 32.43 μL compared with 0 μL at baseline (P < .0001) (Figure 2, C2). Pulmonary venous flow reversal was observed, with systolic to diastolic wave ratio reversal from 0.76 ± 0.18 at baseline to −0.84 ± 1.02 with MR (P < .0001) (Figure 2, C3). MR severity persisted through the entire follow-up (Figure E1). Overall survival in MR group was 92.31% (Figure 2, C4), with 5 rats dying in the first 2 weeks. There were no deaths in the sham group.
Longitudinal LV Chamber Remodeling
Representative images of hearts explanted after 40 weeks are shown in Figure E2, A1-B1, with corresponding B-mode and M-mode frames shown in Figure E2, A2-B3. In the rats with MR, pronounced dilation of the LV chamber is evident, compared with sham (Videos 2 and 3). With MR, EDV increased by 28.36% at 2 weeks (P = .008), 65.01% at 10 weeks (P <.0001), 87.21% at 20 weeks (P <.0001), and 100.17% at 40 weeks (P <.0001), compared with baseline. LV EDV did not change in the sham group (Figure 3, A1). The rate of change in EDV was highest immediately after MR was induced and remained greater than 20 μL/week for the first eight weeks after MR onset (Figure 3, A2). Change in ESV was more gradual—unchanged at 2 weeks, increased by 11.50% at 10 weeks (P < .0001), 126.58% at 20 weeks (P < .0001), and 172.77% at 40 weeks (P<.0001) (Figure 3, B1). The greatest rate of change in ESV occurred after 8 weeks with MR, plateaued by 22 weeks and then increased through 34 weeks (Figure 3, B2). Stroke volume was significantly elevated compared with baseline values at all time points throughout the 40-week study (P <.0001) (Figure 3, C1). After MR onset, EF was elevated compared with baseline values for the first 6 weeks (P <.05) but was otherwise unchanged through 12 weeks compared with baseline (Figure 3, D1). Significant reduction in EF occurred at 14 weeks post-MR (P = .0023) and remained significantly decreased thereafter. By 40 weeks, EF decreased to 56.36 ± 3.06% compared with 66.85 ± 5.15% at baseline (P <.0001). The greatest rate of change in EF occurred at 8 weeks after MR onset and remained negative thereafter (Figure 3, D2).
FIGURE 3.
Longitudinal changes in cardiac function in the sham-operated rats and those induced with severe MR. A1, EDV was significantly elevated in the MR group as early as 4 weeks after inducing the valve lesion and remained significantly elevated throughout the follow-on duration. A2, The rate of change of EDV was greatest in the earliest time period after inducing MR, and plateauing around 8 weeks after inducing the valve lesion. From weeks 8 to 40, the rate of change of EDV was minimal. B1, Similar trends were observed in the changes in ESV, with a rise in ESV occurring at 8 weeks after induction of mitral regurgitation. C1-2, Absolute changes and rate of change of stroke volume. D1-2, Longitudinal changes in ejection fraction, with a significant fall in ejection fraction occurring at 12 weeks after inducing MR. The largest rate of change was also observed within the first 10 weeks and plateaued thereafter. In the graphs, the marker indicates the mean and the shaded region represents the standard deviation. Blue stars below the horizontal axes represent statistical significance when the group means were compared at the same time point (P < .05). Red stars below the horizontal axes indicates statistical significance when comparing the mean value at that time point, to the mean value at baseline in the same group (P < .05). EDV, End-diastolic volume; ESV, end-systolic volume; MR, mitral regurgitation.
LV internal diameter in diastole increased after 2 weeks of MR compared with baseline (P = .022) and remained increased thereafter (Figure E3, A1). LV internal diameter in systole was significantly increased after 8 weeks of MR compared with baseline and remained significantly elevated thereafter (Figure E3, A2). LV anterior wall thickness in diastole slightly increased after MR at 2 weeks compared with sham (P = .032) but remained unchanged at all other time points over 40 weeks (Figure E3, B1). LV posterior wall thickness in diastole and systole was not significantly different compared with sham after onset of MR (Figure E3, C1-C2).
There were no changes in heart rate after MR onset compared with age- and weight-matched shams controls (Table 1). Heart weight was decreased after 2 weeks of MR compared with sham (P < .0001), and then significantly increased at 10, 20, and 40 weeks after MR compared with sham (P = .0011, P = .0068, P < .0001, respectively). When normalized to body weight, heart weight was increased after 2 weeks of MR compared with sham (P < .0001). However, when normalized to tibia length, heart weight was decreased (P = .0001). LV mass and LV mass index followed a similar trend of increasing after onset of MR. LV mass index was significantly greater at 2, 10, and 40 weeks after MR onset, with a slight decrease at 20 weeks, although not significant. Lung weight was significantly higher in the MR groups at 2, 10, and 20 weeks after MR onset compared to age- and weight-matched sham controls (P = .002, P < .0001, P = .0038, respectively).
TABLE 1.
Heart weight, LV mass, and lung weight measured at termination in the MR and sham groups at 2, 10, 20, and 40 weeks
2 wk |
10 wk |
20 wk |
40 wk |
|||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
Sham | MR | P value | Sham | MR | P value | Sham | MR | P value | Sham | MR | P value | |
Heart rate, bpm | 333.50 ± 34.90 | 334.20 ± 25.02 | n.s. | 330.40 ± 26.27 | 314.00 ± 32.23 | n.s. | 319.40 ± 24.40 | 286.70 ± 85.96 | n.s. | 325.90 ± 24.38 | 311.70 ± 44.56 | n.s. |
Heart weight, g | 1.55 ± 0.099 | 1.35 ± 0.10 | .00010 | 1.45 ± 0.10 | 1.65 ± 0.19 | .0011 | 1.52 ± 0.13 | 1.78 ± 0.29 | .0068 | 1.60 ± 0.11 | 2.00 ± 0.36 | .00010 |
Body weight, g | 408.00 ± 22.42 | 404.00 ± 18.82 | n.s. | 445.30 ± 27.74 | 477.30 ± 39.90 | .032 | 450.00 ± 23.20 | 510.70 ± 53.91 | .0006 | 522.70 ± 14.86 | 528.80 ± 51.62 | n.s. |
Tibia length, cm | 4.50 ± 0.23 | 4.41 ± 0.29 | n.s. | 4.62 ± 0.19 | 4.97 ± 0.58 | n.s. | 4.82 ± 0.08 | 4.54 ± 0.40 | .016 | 5.10 ± 0.085 | 4.94 ± 0.25 | .016 |
Heart weight/body weight, % | 0.33 ± 0.027 | 0.38 ± 0.025 | .00010 | 0.33 ± 0.020 | 0.35 ± 0.047 | n.s. | 0.34 ± 0.036 | 0.35 ± 0.048 | n.s. | 0.31 ± 0.019 | 0.38 ± 0.063 | .00030 |
Heart weight/tibia length, g/cm | 0.35 ± 0.036 | 0.30 ± 0.027 | .00010 | 0.32 ± 0.025 | 0.34 ± 0.049 | n.s. | 0.32 ± 0.024 | 0.39 ± 0.076 | .00040 | 0.31 ± 0.022 | 0.40 ± 0.066 | .00010 |
LV mass, mg | 1295.00 ± 144.80 | 1566.00 ± 292.70 | .0033 | 1335.00 ± 187.90 | 1869.00 ± 364.70 | .00010 | 1361.00 ± 147.40 | 1382.00 ± 453.50 | n.s. | 1540.00 ± 150.50 | 1868.00 ± 411.80 | .0070 |
LV mass index | 3.18 ± 0.35 | 3.90 ± 0.84 | .0047 | 2.99 ± 0.31 | 3.94 ± 0.81 | .00020 | 3.00 ± 0.34 | 2.77 ± 0.97 | n.s. | 2.95 ± 0.27 | 3.54 ± 0.72 | .0057 |
Lung weight, g | 2.12 ± 0.61 | 2.95 ± 0.66 | .0020 | 1.81 ± 0.40 | 2.66 ± 0.49 | .00010 | 2.02 ± 0.45 | 2.84 ± 0.54 | .0038 | 2.07 ± 0.54 | 2.45 ± 0.93 | n.s. |
Data are represented as mean ± SD. MR, Mitral regurgitation; n.s., not significant; LV, left ventricle.
LV PV Loops and Myocardial Energetics
Representative PV loops are shown in Figure 4, A. After the onset of MR, we observed widening of the PV loop at 2 weeks (red curve), and a rightward shift by 10 weeks (green curve), 20 weeks (yellow curve), and 40 weeks (cyan curve). PVA and stroke work were significantly increased by 20 and 40 weeks compared with sham (P <.05) (Figure 4, B and C). Potential energy was slightly decreased after 2 weeks of MR compared with sham and significantly increased at 10, 20, and 40 weeks compared with 2 weeks (P = .0259, P = .0108, P < .0001, respectively). By 40 weeks, potential energy was significantly increased compared with sham (P = .0001) (Figure 4, D). The overall efficiency of the LV, the ratio of stroke work and PVA, slowly decreased by 10 weeks after MR and was significantly decreased at 40 weeks compared to sham (P = .0411) (Figure 4, E).
FIGURE 4.
A, Representative pressure–volume loops obtained at the time of termination in each group of animals. The blue line represents the loop from the sham group of animals, the red line indicates the loop at 2 weeks after inducing mitral regurgitation, the green line indicates the loop at 10 weeks after inducing mitral regurgitation, the yellow line indicates the loop at 20 weeks after inducing mitral regurgitation, and the cyan line indicates 40 weeks after inducing mitral regurgitation. A right ward shift was observed in all of the loops from rats with MR compared with the sham group; however, a distinctly large shift was observed in the loops at 20 and 40 weeks. B, The PVA that represents the total mechanical work performed to eject blood in each heartbeat is increased in the MR groups at 20 and 40 weeks compared with the sham group of animals. C, SW, which is the area enclosed within each loop, is representative of the external work done by the ventricle to eject blood into the aorta. SW was increased at 20 and 40 weeks after induction of MR, compared with the sham. D, Depiction of the elastic PE stored in the myocardium at the end of systolic contraction. PE was initially reduced with the onset of MR but rose linearly beyond the initial dip. E, Left ventricular pumping efficiency was significantly reduced at later stages of MR when compared with the sham group. Data are represented as box and whisker plots, the middle line representing the median, the upper and lower borders representing the upper and lower quartiles, and the upper and lower whiskers represent maximum and minimum values. MR, Mitral regurgitation; PVA, pressure volume loop area; SW, stroke work; PE, potential energy.
LV systolic indices derived from pressure volume loops are represented in Figure 5. End-systolic pressure remained unchanged after 2 weeks of MR compared with sham and was significantly decreased at 10 weeks (P = .041) and at 40 weeks (P < .0001) compared with sham (Figure 5, A). dP/dtmax, which represents the maximum rate of pressure rise during isovolumetric contraction, was decreased after 10 weeks of MR compared with sham (P = .0046) but remained unchanged at all other time points (Figure 5, B). End-systolic elastance, which is a more load insensitive measure of LV contractility, was decreased at all time points after the onset of severe MR compared with sham with significance at 10, 20, and 40 weeks (P < .05) (Figure 5, C). Similarly, the relationship between dP/dtmax and EDV, which also indicates a more load insensitive measure of LV contractility compared to dP/dtmax alone, was decreased at all time points after onset of MR compared with sham with significance at 10 and 40 weeks (P<.05) (Figure 5, D). Preload recruitable stroke work was significantly decreased at 40 weeks compared with sham (P = .027) (Figure 5, E). The relationship between PVA and EDV, which represents preload recruitable PVA, also showed a decreasing trend at 10 and 40 weeks after MR onset compared with sham, although not significant (Figure 5, F).
FIGURE 5.
Systolic indices derived from PV loops in the mitral regurgitation (red) and sham (blue). A, ESP, which represents the maximum pressure generated by the left ventricle, was significantly reduced in the mitral regurgitation group at multiple time points compared with the sham animals. B, Maximum rate of change of pressure was relatively equivalent between the groups, except at the 10 week time point. C, Ees, which is an indicator of contractility of the myocardium, was significantly reduced at the 10, 20, and 40 weeks in the mitral regurgitation group compared with the sham group at the same time point. D, Preload adjusted dP/dt max, which is a measure of contractility after correction for preload, was significantly reduced in the mitral regurgitation group at multiple time points. E, PRSW, and F, preload recruitable pressure volume area were not different between the groups. Data are represented as box and whisker plots, where the middle line represents the median, the upper and lower borders represent the upper and lower quartiles, and the upper and lower whiskers represent maximum and minimum values. *P < .05. ESP, End-systolic pressure; Ees, End-systolic elastance; EDV, end-diastolic volume; PRSW, preload recruitable stroke work; PVA, pressure volume loop area.
LV diastolic parameters derived from pressure volume loops are represented in Figure 6. End-diastolic pressure (EDP) showed a biphasic increase from 2 to 40 weeks. After 2 weeks of MR, EDP increased from 8.68 ± 2.10 mm Hg to 10.91 ± 3.83 mm Hg compared with sham. By 10 weeks, EDP was decreased and then subsequently increased to 8.99 ± 1.66 mm Hg by 20 weeks compared with sham (P = .021). By 40 weeks, EDP was unchanged compared with sham (Figure 6, A). The EDP volume relationship did not reveal any significant changes compared with sham (Figure 6, B). dP/dtmin, which represents the maximum pressure decay during isovolumetric relaxation, was significantly decreased after 10 and 40 weeks of MR compared with sham (P = .028, P < .0001, respectively) (Figure 6, C). Tau G was significantly increased after 40 weeks of MR compared with sham (P = .014) (Figure 6, D). Arterial elastance was decreased after onset of MR compared with sham with significance at 40 weeks (P = .0008) (Figure 6, E). Ventriculo-arterial coupling (end-systolic elastance/arterial elastance), which determines the efficiency of energy transfer from the arterial system to systemic circulation, showed a trend of decreasing after 10 weeks of MR onset compared with sham, although none of the values showed significance (Figure 6, F).
FIGURE 6.
Diastolic and ventriculo-arterial coupling in dicesderived from pressure volume loops in the mitral regurgitation (red) and sham (blue) groups. A, EDP that is indicative of the filling pressures or the extent of congestion in the left ventricular chamber demonstrated a biphasic trend. A rise in the EDP was observed initially at 2 weeks due to the severe mitral regurgitation but was reduced thereafter owed to the dilatation of the chamber. The end diastolic pressure was elevated again at 20 weeks, potentially indicating changes in myocardial properties that reduce ventricular distensibility. B, EDPVR and C, minimum dP/dt, which are indicative of diastolic function, were not different between thegroups. D, Tau Glantz(Tau G), E, Ea and F, Ees/Ea were also not different between the groups except at very late stages of 40 weeks. Data are represented as box and whisker plots, where the middle line represents the median, the upper and lower borders represent the upper and lower quartiles, and the upper and lower whiskers represent maximum and minimum values. *P < .05. EDP, Enddiastolic pressure; EDPVR, end-diastolic pressure volume relationship; Ea, arterial elastance; Ees/Ea, ventriculo-arterial elastance ratio.
Longitudinal Transcriptomic Alterations in the LV
RNA-sequencing analysis provided an important glimpse at the key gene signaling events occurring in the LV tissue post-MR. Principle component analysis revealed that the sham and each MR group (2, 10, 20, and 40 weeks) significantly differed (Figure 7). Figure 7 also shows the distribution of the number of significantly altered genes after onset of MR compared with sham. After 2 weeks of MR, 108 genes were up-regulated and 31 genes were down-regulated. After 10 weeks of MR, 73 genes were up-regulated and 47 genes were down-regulated. By 20 weeks, 51 genes were up-regulated and 83 genes were down-regulated. By 40 weeks after MR, 690 genes were up-regulated and 732 genes were down-regulated. Tables E3-E6 represent the top 20 genes that were significantly up-regulated and down-regulated after 2, 10, 20, and 40 weeks of MR compared with sham. Figure 7 and Table E7 shows the top 25 Gene Ontology–enriched terms at each time point. After 2 weeks, there was a significant alteration of the “cell cycle process” and “cytoskeletal part and organization,” suggesting an increase in cell proliferation and changes in the cellular cytoskeletal network. At 10 weeks, there was a significant alteration of “response to stimulus” and “developmental process,” indicating cellular adaptation and activation of fetal gene programming. At 20 weeks, there was a significant alteration of genes belonging to “extracellular region, extracellular matrix structure and constituents” and “response to stimulus,” indicating cellular adaptation response and pathologic extracellular matrix remodeling. By 40 weeks, there was a significant alteration of genes involved in “multicellular organismal process” and “plasma membrane,” suggesting an intricate role of plasma membrane proteins and an organized LV tissue response to MR. Table E8 represents the top 3 Protein Analysis Through Evolutionary Relationships–identified pathways in the up- and down-regulated genes after MR onset.
FIGURE 7.
Volcano plots showing the distribution of significantly altered genes (left), summary of gene expression after MR surgery compared with sham displayed in heat maps (left middle), principle component analysis (right middle), and top 25 Gene Ontology terms (right) in the (A) MR 2-week, (B) MR 10-week, (C) MR 20-week, and (D) MR 40-week groups. MR, Mitral regurgitation.
Kyoto Encyclopedia of Genes and Genomes analysis revealed several biological pathways characterized by the up-regulated and down-regulated genes at each time-point after MR onset, which is summarized in Table E9. After 2 weeks of MR, pathways involving cell cycle, cellular senescence, p53, and renin–angiotensin system signaling were activated. By 10 weeks, the most enriched pathway was the aldosterone synthesis and secretion signaling along with activation of amino-acid metabolism. By 20 weeks, pathways involving regulation of extracellular matrix genes, collagens, elastin and thrombospondins and proinflammatory genes and cytokines (intercellular adhesion molecule 1, vascular cell adhesion molecule 1, and interleukin-17 signaling were activated). By 40 weeks, a plethora of pathways belonging to oxidative phosphorylation, thermogenesis, longevity, AMPK signaling, cardiac muscle contraction, and calcium signaling were activated.
DISCUSSION
Our study is one of the first to map the longitudinal geometric, functional, and transcriptional remodeling in the LV with uncorrected hemodynamic stress from primary MR up to 40 weeks. This duration in the rodent life is equivalent to 20 years in human life, making this study of relevance to the clinical scenario where patients survive with uncorrected primary MR for long periods of time. After the onset of MR, rapid and significant changes occurred in the diastolic geometry and function of the heart, in response to the elevated end-diastolic wall stress from the volume overload. An immediate adaptive response was observed, resulting in an increase in LV EDV by 28% within 2 weeks, 65% within 10 weeks, 87% by 20 weeks, and 100% by 40 weeks. This rise in volumes paralleled reduction in the filling pressures that were acutely elevated after MR onset. LV mass was significantly greater in the MR group at all the time points, indicating a hypertrophic response in the myocardium that occurs early and persists alongside the remodeling process. Mass increased by 18.65% by 2 weeks, 51.5% by 10 weeks, 48.77% by 20 weeks, and 46.68% by 40 weeks, compared with baseline. In the initial 10 weeks, both chamber volume and mass increased, but in the subsequent period only a rise in chamber volume was observed. Change in ventricular geometry and mass would require structural changes in the myocyte or nonmyocyte compartments of the heart. In the early stages after an injury, tissue edema could play a role, but often such edema is resolved within a short period. In the myocyte compartment, Liu and colleagues14 previously demonstrated that cardiomyocytes are deformed in response to volume overload from an arteriovenous fistula, with a rise in both cell length and cross-sectional area. Contrastingly, volume overload in the dog demonstrated elongation but no change in cardiomyocyte cross-sectional area, which may be explained by the lower severity of volume overload in this study.15 Echocardiographic data from this study indicate a rise in ventricular circumference, but not thickness, indicating that cardiomyocyte elongation may have occurred in this model. MR from mitral valve puncture is likely to result in a lower volume overload compared with the arteriovenous fistula, explaining these findings. The nonmyocyte fraction, consisting of vasculature and capillaries, extracellular matrix, fibroblasts, and variety of immune cells, could change with myocardial stress. In a previous investigation in this model, we reported degradation of the extracellular matrix rather than its accumulation, which was corroborated by others in other volume overload models as well.10,16–18 Further investigations in this area are warranted to overcome the current dearth of data.
Systolic function paralleled changes in EDV, with some exceptions. EF was significantly elevated after the onset of MR for 4 weeks, which may be attributed to an acute hyperkinetic response of the LV to increased filling and reduced afterload from the regurgitation. Such a response of the ventricle is observed clinically as well, in patients with acute-onset primary MR from chordal rupture or other lesions. EF was significantly lower at 18 weeks after MR onset when compared with the sham group at the same time point and was lower by 14 weeks when compared with the baseline measurements in the MR group. However, ESV was significantly elevated compared with sham by 10 weeks after MR, and by 8 weeks when compared with the baseline value of the MR group. This is a significant observation that indicates that ESV may be a better and earlier indicator of LV dysfunction. In the 2017 update to the clinical guidelines, mitral valve repair is recommended in patients with severe MR with an EF<60% and a LV ESV greater than 40 mm, as LV dysfunction may have already developed in these patients.6 Our data confirm these findings in the rat, providing the opportunity to study the effect of MR repair in reversing the ventricular remodeling. Despite an early significant rise in ESV, a delayed change in EF may be attributed to the load dependence of this measurement. The afterload reducing effect of MR, uncouples end-systole from end ejection, resulting in greater EFs that are not indicative of cardiac dysfunction. This hypothesis is confirmed from invasive hemodynamic information obtained in this study, where preload adjusted contractile indices were significantly depressed in the regurgitation group by 10 weeks. Such an earlier fall in load-independent contractile indices in the setting of MR have also been reported by Kim and colleagues19 and Rodriguez and colleagues.20
Transcriptomic data from our study provide a critical snapshot of molecular signaling events that take place in the LV upon onset of primary MR in a chronic setting at 4 different time points. Our findings demonstrate that an acute onset of MR (2 weeks) causes differential regulation of genes associated with cellular proliferation, cytoskeletal remodeling, and genes responsible for cellular adaptation to stress signaling. These include genes from nuclear as well as cytoplasmic compartments. Important cell survival and proliferation pathways such as the p53 pathway was activated, which is essential for cellular response to intrinsic and extrinsic stressors; paralleled by up-regulation of several genes required for DNA replication. Several genes in the cardiac thyrotropin-release hormone pathway were upregulated as well, which is a process implicated in cardiac hypertrophy.21 Similar analysis of downregulated genes at this time point revealed involvement of p53 signaling, transcription regulation by bZip transcription factor signaling, endothelin signaling pathway, and Gi alpha– and Gs alpha–mediated pathways, to name a few. p53 signaling is a master regulator of the cardiac transcriptome, which is essential to maintain physiological cardiac tissue homeostasis.22 Elevated wall stress perturbs the native homeostasis in the heart tissue, whose restoration is likely orchestrated by p53 signaling through up-regulation and down-regulation of specific genes. Nomura and colleagues23 recently implicated the role of p53 in early-stage hypertrophy and later stages of heart failure as well, hypothesizing that p53 activation occurs when sustained external stimulus induces accumulative oxidative DNA damage. Interestingly, several genes from Iroquois Homeobox family (Irx1, 3, and 5) were downregulated at 2 weeks post-MR. Irx genes have early regulatory functions in embryonic patterning and specification and play a later role in post development function such as EC coupling, and down-regulation of Irx genes post early onset MR may have many clinical implications.24 These findings warrant better understanding of the mechanisms underlying abnormal conduction in the setting of advanced MR.
By 10 weeks of MR when systolic function was depressed in this model, the most up-regulated signaling cascades were oxidative stress response and angiotensin II stimulated signaling through G proteins and beta-arrestin; and the most downregulated were integrin signaling and Gonadotropin-releasing hormone receptor pathways. Oxidative stress occurs when reactive oxygen species (ROS) are generated by cells and are not adequately countered by the intrinsic antioxidant systems that are intrinsic to most tissues. Such pathologic stress has been implicated in adverse cardiac remodeling and dysfunction in other forms of cardiac stress, including volume overload from different lesions.25,26 Robison and colleages27 recently demonstrated that ROS accumulation in the cardiomyocytes may be governed by intracellular microtubule deformation, which is dependent upon cardiomyocyte resting length. In these dilated hearts with MR, where cardiomyocytes may be elongated, it is possible that such mechano-regulatory accumulation of ROS occurs and warrants further investigation. Previous studies of myocardial tissue from heart failure also demonstrated elevated AngII signaling, which was associated with a rise in ROS generation and oxidative stress.28 Downstream signaling in the AngII pathway is mediated by ROS, likely mediating gene expression that is involved in pathological cardiac remodeling and mitochondrial dysfunction. Gladden and colleagues29 reported in patients with chronic MR, disruption of the mitochondria with parallel rise in ROS. In examining the down-regulated pathways, suppression of integrin signaling is of interest, as chronic hemodynamic stress has been implicated to stimulate this signaling cascade in the heart. Down-regulation of integrin signaling in these hearts may indicate a feedback loop wherein excessive accumulation of associated proteins may have mediated reduced transcription of relevant genes.30 Interestingly, DNA Topoisomerase II alpha (Top2a), which was upregulated at the 2-week time point, was noted to be down-regulated at 10-week time point. The role of Top2a has been well established in the settings of cancer, where it has been shown to play a role in cell proliferation and chemo- and radio-sensitization of tumors.31 Importantly, Top2a knockout mice demonstrate prenatal lethality prior to heart atrial septation, suggesting a critical role of Top2a in MR-based LV pathology.
At 20 weeks, elevated transcription of oxidative stress response genes also paralleled a rise in transforming growth factor-beta signaling, and dopamine receptor–mediated signaling pathways. Transforming growth factor-beta signaling is associated with a variety of signaling cascades, including cardiac hypertrophy and cardiac fibrosis, both of which could occur in these later stages of remodeling.32 The role of dopamine receptor mediated signaling is unclear but may be relevant as distribution of all types of dopamine receptors were detected in the heart in previous studies.33 At this time point, several mechanosensitive pathways, such as integrin, Wnt, and Cadherin signaling, were significantly down-regulated, along with other pathways such as p53, and inflammation mediated by chemokine and cytokine signaling. The dynamics of such suppression must be combined with estimation of proteins in the muscle, which was not within the scope of this work.
By 40 weeks, a new-onset burst of several new genes and pathways were observed, indicating an accelerated momentum toward heart failure. Activation of PI3-kinase and p53 signaling via glucose deprivation signaling indicates endoplasmic reticulum stress, which may indicate cellular metabolic imbalance and apoptosis.34 Significant up-regulation of pathways relevant to adrenaline and noradrenaline biosynthesis, alpha adrenergic receptor signaling, and CCKR signaling at this time point also indicate an adaptation to increased sympathetic activity in the heart. Kaye and colleagues35 previously reported evidence of such elevated cardiac sympathetic activity in heart failure.
We also identified novel long–noncoding RNAs from our study. These include LOC102550325 (down-regulated at 2 weeks post-MR), H19 imprinted maternally expressed transcript (down-regulated at 2 weeks), LOC100910189 (up-regulated at 20 weeks post-MR), and RGD1566401 and LOC102546391 (both down-regulated at 40 weeks post-MR). Although not much is known about the lnc-RNAs identified from our study, H19 has been previously reported to negatively regulate cardiomyocyte remodeling and hypertrophy.36 Collectively, it would be interesting to further elucidate the role of these newly identified noncoding RNAs in the pathophysiology of LV remodeling post-MR.
Advancing these transcriptional changes to clinical application requires further efforts that link gene expression to proteome, proteome to metabolome, and then identifying biomarkers that can be measured in the bloodstream that relate to these changes in the myocardium. In addition, knowledge of the longitudinal transcriptional changes can aid in therapeutic discovery. At 2 weeks, several cytoskeletal remodeling pathways are implicated. Chen and colleagues37 recently demonstrated the role of cytoskeletal remodeling in heart failure progression and demonstrate the benefit of drugs such as parthenolide and colchicine, which modify the detyrosination of the cytoskeletal elements. Transcriptomic data also indicate that the shift from early structural changes to chronic pathologic remodeling is mediated by oxidative stress signaling, which can be targeted with antioxidant drugs.38 In the late stage of remodeling, involvement of several fibrosis and mechano-sensitive genes indicates processes that are similar to other forms of heart failure, unifying later stages of remodeling in MR to other forms of cardiac dysfunction.
Limitations
As with any preclinical study, application of these data to clinical scenarios should be considered within the scope of limitations of a preclinical study. This rodent model of MR has acute onset of severe regurgitation, which represents a subset of patients with degenerative MR. Nonetheless, acute chordal rupture leading to MR is often the most common etiology of patients presenting to surgery. Selection of interrogation windows to assess cardiac dysfunction and transcriptome were arbitrary in this model but were adequately spaced to capture the temporal changes in remodeling in this model. The novel genes identified in this study need to be further examined to determine their functional importance in the context of MR, as well as other cardiac diseases in animal models and patient datasets in the future. Careful examination and subsequent validation of these genes may provide important therapeutic and early diagnostic targets of MR related pathological remodeling of the LV. Finally, although this study reveals a high-resolution, longitudinal map of cardiac remodeling at the functional, geometric and biological levels in the setting of MR, further studies are required to understand the effects of correcting MR at these varied time points to assess the potential for reverse remodeling.
Supplementary Material
VIDEO 1. Video showing the operative procedure of creating mitral regurgitation on the beating heart in the rat, using echocardiographic guidance. Video available at: https://www.jtcvs.org/article/S0022-5223(20)32784-7/fulltext.
VIDEO 2. Video depicting the beating heart in a rat from the sham group, depicting a smaller left atrium and left ventricle. Video available at: https://www.jtcvs.org/article/S0022-5223(20)32784-7/fulltext.
VIDEO 3. Video depicting the beating heart in a rat after 40 weeks of mitral regurgitation, depicting a severely enlarged left atrium and left ventricle. Video available at: https://www.jtcvs.org/article/S0022-5223(20)32784-7/fulltext.
CENTRAL MESSAGE.
Left ventricular dysfunction occurs before fall in ejection fraction in severe mitral regurgitation in an experimental rodent model.
PERSPECTIVE.
Patients with primary mitral regurgitation have preserved ejection fraction, which is currently used as a clinical trigger for intervention. In a rodent model, we demonstrate left ventricular dysfunction and adverse biological remodeling occur before a clinically significant reduction in ejection fraction. These data can be valuable in defining better clinical indices to capture dysfunction.
Acknowledgments
The authors acknowledge Laura Susan Schmarkey for project management support and Robert Hernandez-Merlo, DVM, for veterinary technician support.
This work was funded by American Heart Association grants 14SDG20380081 and 19PRE34380625; National Heart, Lung, and Blood Institute grants R01HL133667, R01HL135145, and R01HL140325; and infrastructure support from the Carlyle Fraser Heart Center at Emory University Hospital Midtown.
Abbreviations and Acronyms
- EDP
end-diastolic pressure
- EDV
end-diastolic volume
- EF
ejection fraction
- ESV
end-systolic volume
- LV
left ventricle/ventricular
- MR
mitral regurgitation
- PV
pressure volume
- PVA
pressure volume loop area
- ROS
reactive oxygen species
- SQ
subcutaneous
- TEE
transesophageal echocardiography
Discussion
Presenter: Dr Daniella Corporan
Dr Amy Hackmann (Dallas, Tex). Thank you, Dr Kane and Dr Ugalde, for inviting me and congratulations on this excellent work, Ms Corporan and Dr Padala. I have a few questions for you. I think the most burning for the audience is: How do we translate this work to clinical care? If we see a patient who maybe is graded only as moderate mitral regurgitation (MR) but has evidence of a dilated left ventricle (LV), should they be referred for early surgery, or we should we still continue to wait for just that severe MR threshold?
Dr Daniella Corporan (Atlanta, Ga). Based on our studies, we see that there’s early and progressive remodeling that occurs very soon after inducing MR. In our case, we see end-diastolic volume increased by 2 weeks, which is equivalent to 1 year in humans. If we look at the current guidelines, which are based off of ejection fraction, we see that the decline in ejection fraction occurs beyond the point at which there’s already active and progressive remodeling. Our data indicate that early surgery could be beneficial, but this needs to be confirmed in larger animals and patients as well.
Dr Hackmann. Thank you. What time frame do we have to intervene on patients? Is our window of opportunity for preventing long-term LV damage a few months, a few years, a decade? What should we recommend for patients?
Dr Corporan. That’s a very interesting question. In our rodent model, we’ve characterized it out to 40 weeks, which is equivalent to about 20 human years. It depends on the indices that we look at. If we look at ejection fraction, it takes a very long time for that parameter to decrease.
But if we look at end-diastolic volume or the rate of change of end-diastolic volume, then those parameters change a lot sooner and so it looks like the 2-week time point, which is equivalent to about 1 year, could be a possible time point to intervene.
Dr Hackmann. Great. So if we start to see LV changes, we should intervene within a year. And since our goal here is to identify relatively asymptomatic patients but who have mitral regurgitation and the potential for LV damage, is there any kind of a screening biomarker or any type of other signal that we can look for in patients other than identifying depressed LV function and severe MR?
Dr Corporan. For the asymptomatic patients, there’s currently no specific biomarker that’s used in the setting of primary MR. But from some of the transcriptomic studies that we’ve shown, we’ve identified some very distinct pathways that are activated at that 2-week time point. In future studies, we want to investigate whether these can be detected as potential biomarkers to be used. In addition to the biomarkers that could be of value, also longitudinal tracing of these patients, possibly with biomarkers, but also with imaging-based LV parameters could be of value as well.
Dr Hackmann. You also showed that there was a very big jump in the end-systolic volume between the 10 and 20 weeks, which would be I guess about a 5-year period in a human patient. Do you know what can account for that? And do we see the same thing if we do longitudinal studies of patients with MR?
Dr Corporan. For that later increase in end-systolic volume, if we think about the LV structurally and what accounts for end-diastolic volume to increase rapidly, we know that cardiomyocytes can elongate to account for some increase in end-diastolic volume. But once that occurs, then that is when possibly the end-systolic volume and the contractile properties of the cardiomyocytes could be altered, which may explain why that occurs a little bit later. With longitudinal tracing, in the future it could be interesting to do some more experiments, potentially on the isolated cardiomyocytes themselves, to look at when exactly the function decreases.
FIGURE E1.
A1, Quantification of regurgitant jet area measured every 2 weeks in the MR (red) and sham (blue) group. A2, Regurgitant jet area normalized to left atrial area. A3, Regurgitant volume. B1, Longitudinal pulmonary venous flow systolic wave S-wave velocity. B2, Pulmonary venous flow systolic wave D-wave velocity. B3, Pulmonary venous flow systolic wave S/D wave ratio. Data are represented as mean ± standard deviation. Red stars represent a statistical significance in the MR group compared with MR baseline values (P < .05). MR, Mitral regurgitation.
FIGURE E2.
A1, and B1, Gross morphology of hearts that had a sham surgery (A1) or MR surgery (B1) after 40 weeks. A2 and B2, Long-axis B-mode images of the left ventricle at 40 weeks. A3 and B3, Long-axis M-mode images of the left ventricle at 40 weeks. MR, Mitral regurgitation.
FIGURE E3.
A, Left ventricular internal diameter at diastole (A1) and systole (A2) measured every 2 weeks in the MR and sham groups. B, Anterior wall thickness at diastole (B1) and systole (B2). C, Posterior wall thickness at diastole (C1) and systole (C2). Data are represented as median and interquartile range. Blue stars represent statistical significance between the MR and sham group at the same time point (P <.05). Red stars represent statistical significance between the MR group and the MR baseline value (P < .05). LV, Left ventricle; MR, mitral regurgitation.
TABLE E1.
Variability among 2 independent users for analysis of TTE LAX B-mode measurements
TTE LAX B-mode |
||||||
---|---|---|---|---|---|---|
End-systolic volume | End-diastolic volume | Stroke volume | Ejection fraction | Fractional shortening | Cardiac output | |
Echo 1 | ||||||
Reviewer 1 | 166.161205 | 524.786137 | 358.624932 | 68.383124 | 17.535455 | 110.368976 |
Reviewer 2 | 110.496628 | 472.261729 | 361.765101 | 76.602672 | 19.424852 | 111.514877 |
% difference | 33.500345 | 10.0087263 | −0.8756137 | −12.019849 | −10.774725 | −1.0382456 |
Echo 2 | ||||||
Reviewer 1 | 169.746356 | 626.68867 | 456.942314 | 72.912136 | 22.011786 | 146.168639 |
Reviewer 2 | 175.280125 | 626.888541 | 451.608416 | 72.039667 | 30.438836 | 142.067967 |
% difference | −3.2600223 | −0.0318932 | 1.16730227 | 1.19660327 | −38.284263 | 2.805439 |
Echo 3 | ||||||
Reviewer 1 | 183.335865 | 548.589935 | 365.25407 | 66.59486 | 15.740376 | 118.718939 |
Reviewer 2 | 185.941383 | 504.093275 | 318.151891 | 63.113695 | 15.292601 | 103.2658 |
% difference | −1.421172 | 8.11109668 | 12.8957301 | 5.22737791 | 2.84475415 | 13.0165744 |
Echo 4 | ||||||
Reviewer 1 | 650.802977 | 1392.80793 | 742.004948 | 53.284405 | 21.484639 | 203.906984 |
Reviewer 2 | 525.542983 | 1288.61826 | 863.075272 | 66.9768 | 22.572024 | 239.212471 |
% difference | 19.2469916 | 7.48054833 | −16.316646 | −25.696815 | −5.0612207 | −17.314506 |
Echo 5 | ||||||
Reviewer 1 | 165.476298 | 561.793798 | 396.3175 | 70.537761 | 21.87421 | 136.632206 |
Reviewer 2 | 174.219272 | 561.496593 | 387.277321 | 68.972337 | 13.93366 | 134.607613 |
% difference | −5.2835204 | 0.05290286 | 2.28104462 | 2.2192709 | 36.3009681 | 1.48178315 |
Echo 6 | ||||||
Reviewer 1 | 177.717098 | 559.953433 | 382.236335 | 68.261732 | 18.860589 | 131.099819 |
Reviewer 2 | 161.593402 | 513.725218 | 352.131816 | 68.544779 | 18.379092 | 119.591938 |
% difference | 9.07267572 | 8.2557249 | 7.8758915 | −0.4146496 | 2.55292663 | 8.77795339 |
Echo 7 | ||||||
Reviewer 1 | 153.665831 | 486.460525 | 332.794694 | 68.372822 | 18.481752 | 103.840034 |
Reviewer 2 | 144.241142 | 417.719898 | 273.478756 | 64.424866 | 15.764034 | 85.445515 |
% difference | 6.13323661 | 14.1307719 | 17.8235828 | 5.7741598 | 14.7048721 | 17.7142845 |
Echo 8 | ||||||
Reviewer 1 | 488.203068 | 1141.73393 | 653.530863 | 57.23981 | 12.382606 | 207.475659 |
Reviewer 2 | 318.241589 | 975.843651 | 657.602063 | 67.388056 | 15.790809 | 206.599099 |
% difference | 34.8136852 | 14.5296794 | −0.6229545 | −17.72935 | −27.524117 | 0.42248811 |
Echo 9 | ||||||
Reviewer 1 | 184.172088 | 550.120803 | 365.948715 | 66.528037 | 15.987639 | 119.394747 |
Reviewer 2 | 217.060877 | 554.904252 | 337.843374 | 60.883184 | 13.068891 | 109.657266 |
% difference | −17.85764 | −0.869527 | 7.68013108 | 8.48492343 | 18.2562791 | 8.15570303 |
Echo 10 | ||||||
Reviewer 1 | 302.808831 | 925.395796 | 622.586965 | 67.290292 | 20.15892 | 179.362154 |
Reviewer 2 | 224.655493 | 971.371345 | 746.715851 | 76.872337 | 23.078151 | 211.480151 |
% difference | 25.8094646 | −4.9682038 | −19.937598 | −14.239862 | −14.481088 | −17.906786 |
Average % difference | 10.0754044 | 5.66998263 | 1.19708698 | −4.719819 | −2.1465614 | 1.6114688 |
TTE, Transthoracic echocardiography.
TABLE E2.
Variability among 2 independent users for analysis of TTE LAX M-mode measurements
TTE LAX M-mode |
||||||||
---|---|---|---|---|---|---|---|---|
IVS; d | IVS; s | LVID; d | LVID; s | LVPW; d | LVPW; s | LV mass | LV mass (corrected) | |
Echo 1 | ||||||||
Reviewer 1 | 1.952557 | 3.299148 | 8.281534 | 5.184375 | 2.221875 | 3.052273 | 1436.90019 | 1149.52015 |
Reviewer 2 | 1.975 | 2.928448 | 8.581034 | 5.380172 | 1.975 | 3.064655 | 1406.65135 | 1125.32108 |
% difference | −1.1494159 | 11.2362343 | −3.6164797 | −3.7766751 | 11.1111111 | −0.4056649 | 2.10514504 | 2.10514501 |
Echo 2 | ||||||||
Reviewer 1 | 2.088506 | 3.246264 | 8.603736 | 5.198563 | 2.752514 | 3.851899 | 1888.45912 | 1510.7673 |
Reviewer 2 | 1.702586 | 2.860345 | 8.717241 | 5.312069 | 2.656034 | 3.95 | 1656.64355 | 1325.31484 |
% difference | 18.4782806 | 11.8880966 | −1.3192525 | −2.1834111 | 3.50515928 | −2.5468217 | 12.2753823 | 12.2753823 |
Echo 3 | ||||||||
Reviewer 1 | 1.885227 | 3.142045 | 8.124432 | 5.049716 | 2.04233 | 3.097159 | 1278.64971 | 1022.91977 |
Reviewer 2 | 1.906897 | 2.928448 | 8.172414 | 5.039655 | 2.451724 | 3.74569 | 1497.24675 | 1197.7974 |
% difference | −1.1494637 | 6.79802485 | −0.590589 | 0.19923893 | −20.045438 | −20.939545 | −17.095929 | −17.095928 |
Echo 4 | ||||||||
Reviewer 1 | 2.173295 | 3.068182 | 12.34375 | 9.034091 | 1.775568 | 2.698864 | 2573.60275 | 2058.8822 |
Reviewer 2 | 1.896552 | 2.758621 | 13.017241 | 9.396552 | 1.982759 | 3.103448 | 2756.8619 | 2205.48952 |
% difference | 12.7337982 | 10.089395 | −5.4561296 | −4.0121469 | −11.668998 | −14.9909 | −7.1207241 | −7.1207241 |
Echo 5 | ||||||||
Reviewer 1 | 2.009052 | 2.894397 | 8.308621 | 5.192888 | 2.309437 | 3.536515 | 1516.05169 | 1212.84136 |
Reviewer 2 | 1.975 | 3.064655 | 8.581034 | 5.175862 | 2.315517 | 3.473276 | 1580.19562 | 1264.1565 |
% difference | 1.69492875 | −5.8823306 | −3.2786789 | 0.3278715 | −0.2632676 | 1.78817282 | −4.2309853 | −4.2309853 |
Echo 6 | ||||||||
Reviewer 1 | 2.334091 | 3.837784 | 8.012216 | 4.690625 | 2.603409 | 3.366477 | 1745.08998 | 1396.07198 |
Reviewer 2 | 2.587931 | 3.813793 | 8.444828 | 4.699138 | 2.519828 | 3.541379 | 1987.00591 | 1589.60473 |
% difference | −10.875326 | 0.62512638 | −5.3994051 | −0.1814897 | 3.21044446 | −5.1954016 | −13.862662 | −13.862662 |
Echo 7 | ||||||||
Reviewer 1 | 2.045455 | 2.769886 | 6.988636 | 4.666193 | 1.768466 | 2.791193 | 967.996317 | 774.397054 |
Reviewer 2 | 1.875 | 2.909483 | 7.306034 | 4.655172 | 2.198276 | 3.103448 | 1140.93557 | 912.748454 |
% difference | 8.3333537 | −5.0398103 | −4.5416302 | 0.23618826 | −24.304114 | −11.187152 | −17.865693 | −17.865693 |
Echo 8 | ||||||||
Reviewer 1 | 2.8125 | 3.853125 | 9.3375 | 6.24375 | 2.5875 | 3.628125 | 2513.26753 | 2010.61402 |
Reviewer 2 | 2.475 | 3.584483 | 9.643966 | 5.803448 | 2.731034 | 4.011207 | 2503.83567 | 2003.06853 |
% difference | 12 | 6.97205515 | −3.2820991 | 7.05188388 | −5.5472077 | −10.558677 | 0.37528277 | 0.37528277 |
Echo 9 | ||||||||
Reviewer 1 | 1.952557 | 2.850284 | 8.057102 | 5.633239 | 1.885227 | 2.423864 | 1221.42243 | 977.137944 |
Reviewer 2 | 1.634483 | 2.656034 | 8.376724 | 5.516379 | 1.77069 | 2.656034 | 1103.21866 | 882.574927 |
% difference | 16.2901262 | 6.81511035 | −3.9669598 | 2.07447261 | 6.07550178 | −9.5785077 | 9.67755038 | 9.6775504 |
Echo 10 | ||||||||
Reviewer 1 | 2.435701 | 3.910606 | 10.105208 | 5.705777 | 2.505533 | 4.106859 | 2500.40103 | 2000.32082 |
Reviewer 2 | 1.994828 | 3.14569 | 10.127586 | 6.291379 | 1.918103 | 2.838793 | 1820.77051 | 1456.61641 |
% difference | 18.1004565 | 19.5600375 | −0.2214502 | −10.263317 | 23.4453108 | 30.8767844 | 27.1808606 | 27.1808607 |
Average % difference | 7.44567387 | 6.30619392 | −3.1672674 | −1.0527385 | −1.4481499 | −4.2737712 | −0.8561772 | −0.8561772 |
TTE, Transthoracic echocardiography; IVS, interventricular septum; d, end diastole; s, end systole; LVID, left ventricular internal dimension; LVPW, left ventricular posterior wall; LV, left ventricular.
TABLE E3.
The top 20 up-regulated and down-regulated genes in the MR 2-week group compared with sham
Up-regulated genes at 2 wk |
Down-regulated genes at 2 wk |
||||||
---|---|---|---|---|---|---|---|
Gene | Description | Fold change | P value | Gene | Description | Fold change | P value |
Polq | DNA Polymerase Theta | 4.67 | .0079 | LOC102550325 | Uncharacterized gene | −4.33 | .023 |
Cmahp | Cytidine Monophospho-N-Acetylneuraminic Acid Hydroxylase, Pseudogene | 4.58 | .043 | Prkar1b | Protein Kinase CAMP-Dependent Type I Regulatory Subunit Beta | −4.21 | .0025 |
Ska1 | Spindle And Kinetochore Associated Complex Subunit 1 | 4.55 | .0020 | Tnnt1 | Troponin T1, Slow Skeletal Type | −3.64 | .028 |
Trh | Thyrotropin Releasing Hormone | 4.52 | .0081 | Irx5 | Iroquois Homeobox 5 | −3.24 | .025 |
Cdk1 | Cyclin Dependent Kinase 1 | 4.17 | .0018 | Irx3 | Iroquois Homeobox 3 | −2.90 | .0079 |
Cdc6 | Cell Division Cycle 6 | 4.14 | .0052 | Cxcl14 | C-X-C Motif Chemokine Ligand 14 | −2.83 | .0049 |
Cenpf | Centromere Protein F | 4.04 | .00021 | Irx1 | Iroquois Homeobox 1 | −2.82 | .032 |
Ccna2 | Cyclin A2 | 3.92 | .0034 | Nfe2 | Nuclear Factor, Erythroid 2 | −2.71 | .034 |
Nek2 | NIMA Related Kinase 2 | 3.88 | .00063 | Ube3d | Ubiquitin Protein Ligase E3D | −2.67 | .027 |
Cdkn3 | Cyclin Dependent Kinase Inhibitor 3 | 3.80 | .0035 | Penk | Proenkephalin | −2.66 | .0031 |
Melk | Maternal Embryonic Leucine Zipper Kinase | 3.70 | .00099 | Hmgcll1 | 3-Hydroxymethyl-3-Methylglutaryl-CoA Lyase Like 1 | −2.60 | .013 |
Ptx3 | Pentraxin 3 | 3.65 | .021 | Epor | Erythropoietin Receptor | −2.56 | .015 |
Pbk | PDZ Binding Kinase | 3.64 | .0022 | Nsg1 | Neuronal Vesicle Trafficking Associated 1 | −2.55 | .011 |
Iqgap3 | IQ Motif Containing GTPase Activating Protein 3 | 3.53 | .00080 | Rec114 | REC114 Meiotic Recombination Protein | −2.51 | .0074 |
Tph1 | Tryptophan Hydroxylase 1 | 3.49 | .022 | Fosb | FosB Proto-Oncogene, AP-1 Transcription Factor Subunit | −2.51 | .014 |
Ccnf | Cyclin F | 3.44 | .00091 | Igfbp3 | Insulin Like Growth Factor Binding Protein 3 | −2.40 | .00051 |
Prc1 | Protein Regulator Of Cytokinesis 1 | 3.40 | .0014 | Cytl1 | Cytokine Like 1 | −2.39 | .046 |
Tpx2 | TPX2 Microtubule Nucleation Factor | 3.38 | .00053 | Msx2 | Msh Homeobox 2 | −2.36 | .020 |
Top2a | DNA Topoisomerase II Alpha | 3.31 | .00036 | Rtn4rl2 | Reticulon 4 Receptor Like 2 | −2.33 | .025 |
Racgap1 | Rac GTPase Activating Protein 1 | 3.30 | .00063 | Cd99l2 | CD99 Molecule Like 2 | −2.31 | .0044 |
TABLE E4.
The top 20 up-regulated and down-regulated genes in the MR 10-week group compared with sham
Up-regulated genes at 10 wk |
Down-regulated genes at 10 wk |
||||||
---|---|---|---|---|---|---|---|
Gene | Description | Fold change | P value | Gene | Description | Fold change | P value |
Ugt1a1 | UDP Glucuronosyltransferase Family 1 Member A1 | 10.91 | .036 | RT1-CE11 | MHC class I family related | −7.63 | .000031 |
Nppa | Natriuretic Peptide A | 5.44 | .029 | Msln | Mesothelin | −5.72 | .0087 |
Dupd1 | Dual Specificity Phosphatase And Pro Isomerase Domain Containing 1 | 5.26 | .0070 | Upk1b | Uroplakin 1B | −5.57 | .037 |
Dusp15 | Dual Specificity Phosphatase 15 | 4.21 | .00077 | Egr3 | Early Growth Response 3 | −4.38 | .0053 |
Pfkfb1 | 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 1 | 3.82 | .000019 | C4a | Complement C4A (Rodgers Blood Group) |
−4.00 | .014 |
Sult1a1 | Sulfotransferase Family 1A Member 1 | 3.33 | .0000072 | Lyc2 | Lysozyme Like 1 | −3.71 | .012 |
Csrp2 | Cysteine And Glycine Rich Protein 2 | 3.10 | .0000044 | Kcna4 | Potassium Voltage-Gated Channel Subfamily A Member 4 | −3.36 | .0014 |
Lrtm2 | Leucine Rich Repeats And Transmembrane Domains 2 | 3.08 | .048 | Cdca3 | Cell Division Cycle Associated 3 | −3.06 | .020 |
Il22ra2 | Interleukin 22 Receptor Subunit Alpha 2 | 3.06 | .0074 | Nipsnap2 | Nipsnap Homolog 2 | −3.05 | .028 |
Tlcd1 | TLC Domain Containing 1 | 2.80 | .035 | Baalc | BAALC Binder Of MAP3K1 And KLF4 | −2.95 | .0041 |
Chrna1 | Cholinergic Receptor Nicotinic Alpha 1 Subunit | 2.79 | .036 | B3gat1 | Beta-1,3-Glucuronyltransferase 1 | −2.76 | .017 |
Oas1k | Oligoadenylate synthetase 1K | 2.74 | .00057 | Top2a | DNA Topoisomerase II Alpha | −2.76 | .0022 |
B3galt5 | Beta-1,3-Galactosyltransferase 5 | 2.71 | .044 | Myo16 | Myosin XVI | −2.69 | .00054 |
Per2 | Period Circadian Regulator 2 | 2.60 | .0000050 | H19 | H19 Imprinted Maternally Expressed Transcript | −2.67 | .046 |
Hpd | 4-Hydroxyphenylpyruvate Dioxygenase | 2.60 | .00018 | Hmgcll1 | 3-Hydroxymethyl-3-Methylglutaryl-CoA Lyase Like 1 | −2.55 | .0064 |
Bex1 | Brain Expressed X-Linked 1 | 2.59 | .0020 | Col1a1 | Collagen Type I Alpha 1 Chain | −2.51 | .011 |
Sdcbp2 | Syndecan Binding Protein 2 | 2.59 | .0073 | Basp1 | Brain Abundant Membrane Attached Signal Protein 1 | −2.46 | .0047 |
Cplx1 | Complexin 1 | 2.56 | .010 | Fosb | FosB Proto-Oncogene, AP-1 Transcription Factor Subunit | −2.44 | .011 |
Agt | Angiotensinogen | 2.55 | .0070 | Ldlr | Low Density Lipoprotein Receptor | −2.44 | .0016 |
Tnfrsf25 | TNF Receptor Superfamily Member 25 | 2.51 | .000029 | Cyr61 | Cellular Communication Network Factor 1 | −2.43 | .00073 |
TABLE E5.
The top 20 up-regulated and down-regulated genes in the MR 20-week group compared with sham
Up-regulated genes at 20 wk |
Down-regulated genes at 20 wk |
||||||
---|---|---|---|---|---|---|---|
Gene | Description | Fold change | P value | Gene | Description | Fold change | P value |
Dupd1 | Dual Specificity Phosphatase And Pro Isomerase Domain Containing 1 |
10.09 | .0000063 | Ptx3 | Pentraxin 3 | −7.48 | .040 |
Adra2c | Adrenoceptor Alpha 2C | 6.62 | .0061 | Upk1b | Uroplakin 1B | −5.13 | .040 |
Ephx4 | Epoxide Hydrolase 4 | 5.50 | .023 | Msln | Mesothelin | −4.80 | .015 |
Dusp15 | Dual Specificity Phosphatase 15 | 4.70 | .0000016 | Cdca3 | Cell Division Cycle Associated 3 | −4.08 | .010 |
Atp1a3 | ATPase Na+/K+ Transporting Subunit Alpha 3 | 4.58 | .0012 | Ccl2 | C-C Motif Chemokine Ligand 2 | −3.70 | .019 |
LOC100910189 | Long noncoding RNA related gene | 4.23 | .012 | Wnt9b | Wnt Family Member 9B | −3.67 | .016 |
Camk2b | Calcium/Calmodulin Dependent Protein Kinase II Beta | 4.17 | .000090 | Robo3 | Roundabout Guidance Receptor 3 | −3.66 | .042 |
Sox8 | SRY-Box Transcription Factor 8 | 3.96 | .0099 | Krt5 | Keratin 5 | −3.63 | .046 |
Sypl2 | Synaptophysin Like 2 | 3.84 | .0014 | Egr2 | Early Growth Response 2 | −3.45 | .015 |
Chrna1 | Cholinergic Receptor Nicotinic Alpha 1 Subunit | 3.82 | .00091 | Col1a1 | Collagen Type I Alpha 1 Chain | −3.41 | .0010 |
Cds1 | CDP-Diacylglycerol Synthase 1 | 3.30 | .00028 | Egr3 | Early Growth Response 3 | −3.40 | .0057 |
Maoa | Monoamine Oxidase A | 3.28 | .00025 | Scara3 | Scavenger Receptor Class A Member 3 | −3.38 | .036 |
Fa2h | Fatty Acid 2-Hydroxylase | 3.19 | .0025 | C4a | Complement C4A (Rodgers Blood Group) | −3.34 | .041 |
Pfkfb1 | 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 1 | 3.18 | .00026 | Has1 | Hyaluronan Synthase 1 | −3.20 | .033 |
Oas1k | Oligoadenylate synthetase 1K | 3.17 | .00042 | Slc1a5 | Solute Carrier Family 1 Member 5 | −3.13 | .00072 |
Epn3 | Epsin 3 | 3.16 | .00019 | Pcdh17 | Protocadherin 17 | −3.09 | .017 |
Hpd | 4-Hydroxyphenylpyruvate Dioxygenase | 3.10 | .0011 | Pimreg | PICALM Interacting Mitotic Regulator | −3.09 | .015 |
Habp2 | Hyaluronan Binding Protein 2 | 3.00 | .0035 | Adamts3 | ADAM Metallopeptidase With Thrombospondin Type 1 Motif 3 | −3.07 | .022 |
Gdf15 | Growth Differentiation Factor 15 | 2.99 | .00077 | Fosb | FosB Proto-Oncogene, AP-1 Transcription Factor Subunit | −3.03 | .0031 |
Ankrd23 | Ankyrin Repeat Domain 23 | 2.97 | .000027 | Thbs1 | Thrombospondin 1 | −3.01 | .00048 |
TABLE E6.
The top 20 up-regulated and down-regulated genes in the MR 40-week group compared with sham
Up-regulated genes at 40 wk |
Down-regulated genes at 40 wk |
||||||
---|---|---|---|---|---|---|---|
Gene | Description | Fold change | P value | Gene | Description | Fold change | P value |
Adcy1 | Adenylate Cyclase 1 | 17.88 | .00000028 | Cldn11 | Claudin 11 | −11.15 | .027 |
Syt13 | Synaptotagmin 13 | 17.19 | .00035 | Cdca3 | Cell Division Cycle Associated 3 | −7.47 | .00096 |
Nos1 | Nitric Oxide Synthase 1 | 14.39 | .000026 | Postn | Periostin | −6.55 | .036 |
Tgfa | Transforming Growth Factor Alpha | 13.94 | .0000032 | Zg16 | Zymogen Granule Protein 16 | −5.75 | .040 |
Fbxo32 | F-Box Protein 32 | 13.41 | .0000000076 | RGD1566401 | Maternally Expressed 3 like | −5.41 | .00011 |
Adra2c | Adrenoceptor Alpha 2C | 12.74 | .00018 | Cblc | Cbl Proto-Oncogene C | −4.99 | .000038 |
Atp1a3 | ATPase Na+/K+ Transporting Subunit Alpha 3 | 11.78 | .000038 | Tnnt1 | Troponin T1, Slow Skeletal Type | −4.90 | .0042 |
Hcn4 | Hyperpolarization Activated Cyclic Nucleotide Gated Potassium Channel 4 | 10.48 | .00016 | LOC102546391 | Long noncoding RNA like gene | −4.84 | .000010 |
Tbx20 | T-Box Transcription Factor 20 | 9.80 | .0000016 | Lpal2 | Lipoprotein(A) Like 2, Pseudogene | −4.59 | .040 |
Ccnd2 | Cyclin D2 | 9.69 | .00000099 | Fscn3 | Fascin Actin-Bundling Protein 3 | −4.33 | .00040 |
Maoa | Monoamine Oxidase A | 9.62 | .0000036 | Myl6b | Myosin Light Chain 6B | −4.33 | .00011 |
Prkaa2 | Protein Kinase AMP-Activated Catalytic Subunit Alpha 2 | 8.23 | .0000057 | Snrpg | Small Nuclear Ribonucleoprotein Polypeptide G | −4.20 | .00018 |
Dgkg | Diacylglycerol Kinase Gamma | 7.86 | .00018 | Kif22 | Kinesin Family Member 22 | −4.13 | .00017 |
Cavin4 | Caveolae Associated Protein 4 | 7.56 | .0000039 | Col11a2 | Collagen Type XI Alpha 2 Chain | −4.12 | .0000043 |
Cds1 | CDP-Diacylglycerol Synthase 1 | 7.22 | .00000073 | Psme2 | Proteasome Activator Subunit 2 | −4.11 | .0000014 |
Gfod1 | Glucose-Fructose Oxidoreductase Domain Containing 1 | 7.06 | .0000015 | Zmynd10 | Zinc Finger MYND-Type Containing 10 | −4.03 | .0000031 |
Ncam1 | Neural Cell Adhesion Molecule 1 | 6.92 | .0000017 | RT1-CE14 | MHC class I family related | −4.01 | .00000076 |
Dupd1 | Dual Specificity Phosphatase And Pro Isomerase Domain Containing 1 | 6.76 | .00020 | Cdca8 | Cell Division Cycle Associated 8 | −3.97 | .00061 |
Pkia | CAMP-Dependent Protein Kinase Inhibitor Alpha | 6.73 | .0000080 | Pimreg | PICALM Interacting Mitotic Regulator | −3.95 | .0079 |
Prkn | Parkin RBR E3 Ubiquitin Protein Ligase | 6.68 | .000024 | Syndig1 | Synapse Differentiation Inducing 1 | −3.88 | .00058 |
TABLE E7.
GO analysis of top 25 significantly altered categories, comparing MR versus sham differentially expressed genes
Category | GO-BP term | Enrichment score | Gene count | Gene symbol | P value |
---|---|---|---|---|---|
MR 2 wk | |||||
GO:0022402 | Cell cycle process | 54.41 | 44 | NEK2, NUF2, KNSTRN, INCENP, CKAP2, BUB1B, TOP2A, RACGAP1, KIF18B, KIF20A, NDC80, KIF11, KIF22, KIF23, KIF2C, SGO2, KIFC1, TPX2, ESPL1, E2F1, E2F7, CIT, IQGAP3, TACC3, ASPM, CDC6, CCNF, CDK1, KNTC1, CKS2, ECT2, FOXM1, NUSAP1, BIRC5, SPC25, MYBL2, CDC20, CCNA2, CCNB1, AURKB, CDKN3, CEP55, UBE2C, CENPW | 2.34E-24 |
GO:1903047 | Mitotic cell cycle process | 62.82 | 40 | NEK2, NUF2, KNSTRN, INCENP, CKAP2, BUB1B, TOP2A, RACGAP1, KIF18B, KIF20A, NDC80, KIF11, KIF22, KIF23, KIF2C, KIFC1, TPX2, ESPL1, E2F1, E2F7, CIT, IQGAP3, TACC3, CDC6, CCNF, CDK1, KNTC1, CKS2, ECT2, FOXM1, NUSAP1, BIRC5, SPC25, MYBL2, CDC20, CCNA2, CCNB1, AURKB, CEP55, UBE2C | 5.21E-28 |
GO:0044430 | Cytoskeletal part | 24.22 | 41 | ACKR2, NEK2, KNSTRN, INCENP, CKAP2, TNNT1, BUB1B, TOP2A, RACGAP1, KIF18B, KIF20A, KIF20B, SARM1, PRC1, NDC80, KIF11, KIF22, KIF23, KIF2C, KIFC1, SKA1, TPX2, ESPL1, E2F1, TACC3, ASPM, CDC6, CCNF, CDK1, KNTC1, ECT2, NUSAP1, BIRC5, CDC20, HMMR, CCNB1, CCNB2, AURKB, LMNB1, CEP55, CENPF | 3.04E-11 |
GO:0051726 | Regulation of cell cycle | 34.15 | 39 | NEK2, MSX2, KNSTRN, INCENP, MKI67, BUB1B, TOP2A, RACGAP1, KIF20A, KIF20B, PRC1, NDC80, KIF11, KIF23, SGO2, TGM1, TPX2, E2F1, E2F7, CIT, TACC3, ASPM, CDC6, CCNF, CDK1, KNTC1, CKS2, ECT2, FOXM1, NUSAP1, BIRC5, CDC20, CCNA2, CCNB1, CCNB2, AURKB, CDKN3, UBE2C, CENPF | 1.48E-15 |
GO:0010564 | Regulation of cell cycle process | 36.06 | 32 | NEK2, MSX2, KNSTRN, INCENP, MKI67, BUB1B, RACGAP1, KIF20A, KIF20B, PRC1, NDC80, KIF11, KIF23, SGO2, TPX2, E2F1, E2F7, CIT, TACC3, CDC6, CCNF, CDK1, ECT2, FOXM1, NUSAP1, BIRC5, CDC20, CCNB1, CCNB2, AURKB, UBE2C, CENPF | 2.18E-16 |
GO:0007010 | Cytoskeleton organization | 24.06 | 30 | NEK2, DIAPH3, NUF2, KNSTRN, TNNT1, RACGAP1, KIF18B, KIF20A, PRC1, NDC80, KIF11, KIF23, KIF2C, KIFC1, TPX2, ESPL1, CIT, TACC3, ASPM, CCL7, CDK1, NUSAP1, BIRC5, SPC25, MYBL2, CDC20, HMMR, CCNB1, AURKB, CENPW | 3.56E-11 |
GO:0007017 | Microtubule-based process | 27.45 | 27 | NEK2, NUF2, KNSTRN, RACGAP1, KIF18B, KIF20A, KIF20B, PRC1, NDC80, KIF11, KIF22, KIF23, KIF2C, KIFC1, TPX2, ESPL1, TACC3, ASPM, CDK1, NUSAP1, BIRC5, SPC25, MYBL2, CDC20, CCNB1, AURKB, CENPW | 1.20E-12 |
GO:0000226 | Microtubule cytoskeleton Organization | 29.75 | 23 | NEK2, NUF2, KNSTRN, KIF18B, KIF20A, PRC1, NDC80, KIF11, KIF2C, KIFC1, TPX2, ESPL1, TACC3, ASPM, CDK1, NUSAP1, BIRC5, SPC25, MYBL2, CDC20, CCNB1, AURKB, CENPW | 1.20E-13 |
GO:0005819 | Spindle | 38.86 | 21 | KNSTRN, INCENP, BUB1B, RACGAP1, KIF20A, PRC1, KIF11, KIF22, KIF23, KIFC1, SKA1, TPX2, ESPL1, TACC3, ASPM, CDC6, CDK1, ECT2, NUSAP1, AURKB, CENPF | 1.32E-17 |
GO:0051301 | Cell division | 29.4 | 20 | NUF2, KNSTRN, TOP2A, KIF18B, KIF20A, KIF2C, KIFC1, SKA1, ASPM, CDC6, CCNF, CDK1, CKS2, BIRC5, SPC25, CDC20, CDCA2, CCNB1, CENPT, CENPW | 1.71E-13 |
GO:0007051 | Spindle organization | 37.77 | 18 | NEK2, NUF2, KNSTRN, KIF20A, NDC80, KIF11, KIFC1, TPX2, ESPL1, TACC3, ASPM, CDK1, SPC25, MYBL2, CDC20, CCNB1, AURKB, CENPW | 3.97E-17 |
GO:0007059 | Chromosome Segregation | 40.86 | 18 | NEK2, NUF2, KNSTRN, INCENP, T0P2A, KIF18B, HJURP, NDC80, SKA1, ESPL1, CIT, NUSAP1, BIRC5, SPC25, CDCA2, CENPF, CENPT, CENPW | 1.80E-18 |
GO:0051276 | Chromosome organization | 22.63 | 18 | NEK2, NUF2, KNSTRN, INCENP, BUB1B, T0P2A, KIF18B, NDC80, KIF22, SG02, ESPL1, CIT, CDK1, NUSAP1, CDC20, CENPI, CENPT, CENPW | 1.49E-10 |
GO:0090068 | Positive Regulation of cell cycle process | 22.72 | 17 | MSX2, RACGAP1, KIF20B, NDC80, KIF23, SG02, E2F7, CIT, CDC6, CDK1, ECT2, NUSAP1, BIRC5, CDC20, CCNB1, AURKB, UBE2C | 1.36E-10 |
GO:0000776 | Kinetochore | 36.17 | 17 | NEK2, NUF2, KNSTRN, INCENP, BUB1B, HJURP, NDC80, KIF2C, SG02, MIS18BP1, BIRC5, SPC25, AURKB, CENPF, CENPI, CENPT, CENPW | 1.96E-16 |
GO:0051783 | Regulation of Nuclear division | 26.71 | 17 | NEK2, MSX2, MKI67, BUB1B, KIF20B, NDC80, KIF11, SG02, CIT, TACC3, CDC6, NUSAP1, BIRC5, CDC20, CCNB1, AURKB, UBE2C | 2.52E-12 |
GO:0051983 | Regulation of chromosome segregation | 30.92 | 15 | NEK2, KNSTRN, MKI67, BUB1B, RACGAP1, NDC80, KIF2C, SG02, CIT, TACC3, CDC6, ECT2, BIRC5, CCNB1, AURKB | 3.74E-14 |
GO:0030496 | Midbody | 25.8 | 15 | NEK2, INCENP, RACGAP1, KIF20A, KIF20B, PRC1, KIF23, CIT, ASPM, CDK1, ECT2, BIRC5, AURKB, CEP55, CENPF | 6.27E-12 |
GO:0072686 | Mitotic spindle | 28.32 | 13 | KNSTRN, RACGAP1, KIF11, KIF22, KIF23, KIFC1, SKA1, TPX2, ESPL1, TACC3, CDK1, ECT2, NUSAP1 | 5.03E-13 |
GO:1902850 | Microtubule cytoskeleton organization involved in mitosis | 25.48 | 13 | NEK2, NUF2, NDC80, KIF11, KIFC1, TPX2, TACC3, NUSAP1, SPC25, MYBL2, CDC20, CCNB1, AURKB | 8.62E-12 |
GO:0007052 | Mitotic spindle organization | 27 | 12 | NEK2, NUF2, NDC80, KIF11, KIFC1, TPX2, TACC3, SPC25, MYBL2, CDC20, CCNB1, AURKB | 1.88E-12 |
GO:0000775 | Chromosome, centromeric region | 26.49 | 11 | INCENP, MKI67, KIF20A, HJURP, KIF2C, SG02, BIRC5, AURKB, CENPF, CENPT, CENPW | 3.13E-12 |
GO:0000777 | Condensed chromosome kinetochore | 27.01 | 11 | NUF2, KNSTRN, BUB1B, HJURP, NDC80, KIF2C, MIS18BP1, BIRC5, SPC25, CENPT, CENPW | 1.86E-12 |
GO:0032465 | Regulation of cytokinesis | 22.55 | 11 | INCENP, RACGAP1, KIF20A, KIF20B, PRC1, KIF23, E2F7, CIT, CDC6, ECT2, AURKB | 1.61E-10 |
GO:0000281 | Mitotic cytokinesis | 22.93 | 10 | INCENP, CKAP2, RACGAP1, KIF20A, KIF23, CIT, ECT2, NUSAP1, BIRC5, CEP55 | 1.10E-10 |
MR 10 wk | |||||
GO:0032502 | Developmental Process | 7.54 | 52 | ARHGAP5, C0L1A1, KCNK2, UGT1A1, MSLN, RH0BTB1, MSX2, NPFF, NPPA, NRP2, PER2, MKI67, T0P2A, PNMT, PLA2R1, SYPL2, TCAP, S0X4, HIVEP3, BASP1, AGT, H19, HSD17B1, PLAGL1, PFKFB1, FANCD2, CSRP2, UPK1B, SLC12A5, FA2H, DMP1, EGR3, CYS1, TDRD1, LIMS1, EYA2, CAMK2B, GLI1, GPC3, CHRNA1, MY016, CCNB2, CCND2, AURKB, WDR62, MEGF9, JPH2, DPYSL3, LDLR, LIM2. SSTR3. ATP1A3 | 5.29E-04 |
GO:0050896 | Response to stimulus | 6.39 | 52 | COL1A1, ACKR2, LYC2, KCNK2, UGT1A1, MSX2, NPFF, NPPA, NRP2, PER2, OPN4, MKI67, TOP2A, PNMT, PLA2R1, ANKRD23, TCAP, SOX4, C4A, CPEB1, AGT, H19, CFD, B3GAT1, HSD17B1, PFKFB1, OAS1K, TAOK3, FANCD2, UPK1B, CNR2, SLC12A5, SULT1A1, EGR3, FCNA, CASTOR1, LIMS1, CAMK2B, FOSB, GLI1, CARD9, GPC3, B3GALT5, CHRNA1, CCND2, AURKB, DPYSL3, LDLR, MC4R, PANX2, SSTR3, ATP1A3 | 1.68E-03 |
GO:0048856 | Anatomical structure development | 7.06 | 39 | ARHGAP5, COL1A1, KCNK2, UGT1A1, MSLN, MSX2, NPFF, NPPA, NRP2, MKI67, TOP2A, PNMT, SYPL2, TCAP, SOX4, AGT, H19, HSD17B1, PFKFB1, CSRP2, SLC12A5, DMP1, EGR3, CYS1, TDRD1, LIMS1, EYA2, GLI1, GPC3, MYO16, CCNB2, CCND2, WDR62, MEGF9, JPH2, DPYSL3, LIM2, SSTR3, ATP1A3 | 8.59E-04 |
GO:0006950 | Response to stress | 6.93 | 35 | COL1A1, ACKR2, LYC2, KCNK2, UGT1A1, MSX2, NPFF, NPPA, PER2, MKI67, TOP2A, PNMT, PLA2R1, TCAP, SOX4, C4A, CPEB1, AGT, H19, CFD, B3GAT1, PFKFB1, OAS1K, TAOK3, FANCD2, CNR2, SLC12A5, FCNA, CAMK2B, GLI1, CARD9, DPYSL3, LDLR, PANX2, SSTR3 | 9.76E-04 |
GO:0009605 | Response to external stimulus | 7.39 | 26 | COL1A1, LYC2, KCNK2, UGT1A1, NPPA, PER2, OPN4, ANKRD23, TCAP, C4A, CPEB1, AGT, H19, CFD, PFKFB1, OAS1K, UPK1B, CNR2, FCNA, CASTOR1, FOSB, CARD9, GPC3, B3GALT5, LDLR, SSTR3 | 6.19E-04 |
GO:0048513 | Animal organ development | 6.7 | 24 | ARHGAP5, COL1A1, UGT1A1, MSLN, MSX2, NPFF, NPPA, NRP2, MKI67, TOP2A, PNMT, SYPL2, TCAP, SOX4, AGT, H19, HSD17B1, PFKFB1, CYS1, GLI1, GPC3, CCNB2, CCND2, LIM2 | 1.24E-03 |
GO:0009719 | Response to endogenous stimulus | 5.82 | 21 | COL1A1, UGT1A1, MSX2, NPPA, TOP2A, PNMT, CPEB1, AGT, H19, PFKFB1, SULT1A1, EGR3, CASTOR1, LIMS1, CAMK2B, FOSB, CCND2, LDLR, MC4R, SSTR3, ATP1A3 | 2.96E-03 |
GO:0009628 | Response to abiotic stimulus | 6.82 | 21 | COL1A1, KCNK2, NPPA, PER2, OPN4, MKI67, TOP2A, PNMT, ANKRD23, TCAP, SOX4, CPEB1, AGT, B3GAT1, FANCD2, SLC12A5, FOSB, CCND2, AURKB, LDLR, ATP1A3 | 1.10E-03 |
GO:0009725 | Response to hormone | 6.01 | 17 | COL1A1, UGT1A1, MSX2, NPPA, TOP2A, PNMT, CPEB1, AGT, H19, PFKFB1, SULT1A1, FOSB, CCND2, LDLR, MC4R, SSTR3, ATP1A3 | 2.47E-03 |
GO:1901652 | Response to peptide | 6.74 | 12 | COL1A1, NPPA, PNMT, CPEB1, AGT, H19, PFKFB1, CARD9, CCND2, LDLR, MC4R, ATP1A3 | 1.18E-03 |
GO:0043434 | Response to peptide hormone | 5.87 | 10 | COL1A1, NPPA, PNMT, CPEB1, AGT, H19, PFKFB1, CCND2, LDLR, MC4R | 2.83E-03 |
GO:0031960 | Response to corticosteroid | 5.99 | 8 | COL1A1, UGT1A1, PNMT, H19, PFKFB1, SULT1A1, FOSB, SSTR3 | 2.49E-03 |
GO:0022836 | Gated channel activity | 5.94 | 7 | KCNA4, KCNK2, KCNN3, TMEM63C, KCNJ14, CHRNA1, JPH2 | 2.64E-03 |
GO:0022839 | Ion-gated channel activity | 6.08 | 7 | KCNA4, KCNK2, KCNN3, TMEM63C, KCNJ14, CHRNA1, JPH2 | 2.28E-03 |
GO:0006813 | Potassium ion transport | 7.2 | 6 | KCNA4, KCNK2, KCNN3, KCNJ14, SLC12A5, ATP1A3 | 7.48E-04 |
GO:0015079 | Potassium ion transmembrane transporter activity | 7.53 | 6 | KCNA4, KCNK2, KCNN3, KCNJ14, SLC12A5, ATP1A3 | 5.35E-04 |
GO:0071805 | Potassium ion transmembrane transport | 7.79 | 6 | KCNA4, KCNK2, KCNN3, KCNJ14, SLC12A5, ATP1A3 | 4.14E-04 |
GO:0006956 | Complement activation | 6.29 | 3 | C4A, CFD, FCNA | 1.85E-03 |
GO:0044305 | Calyx of Held | 6.79 | 3 | KCNK2, CPLX1, ATP1A3 | 1.13E-03 |
GO:0006584 | Catecholamine metabolic process | 5.97 | 3 | PNMT, SULT1A1, MAOA | 2.55E-03 |
GO:0009712 | Catechol-containing compound metabolic Process | 5.97 | 3 | PNMT, SULT1A1, MAOA | 2.55E-03 |
GO:0042923 | Neuropeptide binding | 6.31 | 2 | MC4R, SSTR3 | 1.82E-03 |
GO:0008499 | UDP-Galactose:beta-N-acetylglucosamine beta-1,3-Galactosyltransferase activity | 6.31 | 2 | B3GAT1, B3GALT5 | 1.82E-03 |
GO:0005184 | Neuropeptide hormone activity | 6.03 | 2 | NPFF, NPPA | 2.41E-03 |
GO:0090427 | Activation of meiosis | 7.54 | 2 | MSX2, CAMK2B | 5.30E-04 |
MR 20 wk | |||||
GO:0050896 | Response to stimulus | 9.02 | 62 | COL1A1, COL1A2, LOXL2, COL3A1, LYC2, KCNE1, NPPA, PDK4, OPN4, MKI67, PLLP, TOP2A, PNMT, ICAM1, NCAM1, PRCP, PTX3, KIF22, ANKRD23, SQLE, UCP3, WNT9B, C4A, MPEG1, VCAM1, H19, ELN, FN1, LCK, FBXO32, NES, NPY, IQGAP3, PFKFB1, ATF3, ASS1, DISC1, CCL2, OAS1K, FANCD2, UPK1B, FOSL1, SULT1A1, NR4A1, FOXC2, EGR2, EGR3, RASGRP4, HABP2, PRR5L, CAMK2B, FOSB, HAS1, CARD9, CHRNA1, THBS1, HDAC9, PTPRT, CCNB1, REEP6, LDLR, ATP1A3 | 1.21E-04 |
GO:0009605 | Response to external stimulus | 13.57 | 35 | COL1A1, COL3A1, LYC2, NPPA, PDK4, OPN4, ICAM1, NCAM1, PTX3, ANKRD23, UCP3, WNT9B, C4A, MPEG1, VCAM1, H19, ELN, FN1, LCK, NES, NPY, PFKFB1, ATF3, ASS1, CCL2, OAS1K, UPK1B, FOSL1, RASGRP4, FOSB, CARD9, THBS1, CCNB1, REEP6, LDLR | 1.28E-06 |
GO:0009719 | Response to endogenous stimulus | 12.2 | 30 | COL1A1, COL1A2, COL3A1, KCNE1, NPPA, TOP2A, PNMT, ICAM1, UCP3, VCAM1, H19, ELN, FN1, FBXO32, PFKFB1, ASS1, CCL2, FOSL1, SULT1A1, NR4A1, FOXC2, EGR2, EGR3, CAMK2B, FOSB, HAS1, THBS1, HDAC9, LDLR, ATP1A3 | 5.03E-06 |
GO:0009628 | Response to abiotic stimulus | 11.67 | 28 | COL1A1, LOXL2, COL3A1, KCNE1, NPPA, OPN4, MKI67, TOP2A, PNMT, ICAM1, ANKRD23, UCP3, VCAM1, ELN, LCK, FBXO32, NES, NPY, DISC1, CCL2, FANCD2, FOSL1, FOSB, THBS1, CCNB1, REEP6, LDLR, ATP1A3 | 8.52E-06 |
GO:0044421 | Extracellular region part | 11.25 | 26 | COL1A1, COL1A2, LOXL2, COL3A1, MSLN, COL8A1, NPPA, ADAMTS3, LRRC17, ICAM1, PTX3, GPRC5A, WNT9B, TPX2, C4A, VCAM1, ELN, FN1, GDF15, NPY, CCL2, HABP2, SCARA3, THBS1, CDCA8, LDLR | 1.29E-05 |
GO:1901698 | Response to nitrogen compound | 10.9 | 26 | COL1A1, COL1A2, COL3A1, KCNE1, NPPA, PNMT, ICAM1, NCAM1, UCP3, VCAM1, H19, FN1, PFKFB1, ASS1, CCL2, FOSL1, NR4A1, EGR2, HABP2, CAMK2B, FOSB, CARD9, THBS1, HDAC9, LDLR, ATP1A3 | 1.85E-05 |
GO:0010243 | Response to organonitrogen compound | 10.58 | 25 | COL1A1, COL1A2, COL3A1, KCNE1, NPPA, PNMT, ICAM1, NCAM1, UCP3, VCAM1, H19, FN1, PFKFB1, ASS1, CCL2, FOSL1, NR4A1, EGR2, HABP2, CAMK2B, FOSB, CARD9, HDAC9, LDLR, ATP1A3 | 2.54E-05 |
GO:0009725 | Response to hormone | 11.51 | 24 | COL1A1, COL1A2, NPPA, TOP2A, PNMT, ICAM1, UCP3, H19, ELN, FN1, FBXO32, PFKFB1, ASS1, CCL2, FOSL1, SULT1A1, NR4A1, FOXC2, EGR2, FOSB, THBS1, HDAC9, LDLR, ATP1A3 | 1.01E-05 |
GO:0005615 | Extracellular space | 10.7 | 22 | COL1A1, COL1A2, LOXL2, COL3A1, MSLN, COL8A1, NPPA, ADAMTS3, LRRC17, ICAM1, PTX3, WNT9B, C4A, VCAM1, ELN, FN1, GDF15, NPY, CCL2, HABP2, SCARA3, THBS1 | 2.25E-05 |
GO:1901652 | Response to peptide | 13.38 | 18 | COL1A1, NPPA, PNMT, ICAM1, UCP3, VCAM1, H19, FN1, PFKFB1, ASS1, CCL2, NR4A1, EGR2, HABP2, CARD9, HDAC9, LDLR, ATP1A3 | 1.55E-06 |
GO:0031960 | Response to corticosteroid | 14.68 | 14 | COL1A1, PNMT, ICAM1, UCP3, H19, ELN, FN1, FBXO32, PFKFB1, ASS1, CCL2, FOSL1, SULT1A1, FOSB | 4.20E-07 |
GO:0043434 | Response to peptide hormone | 9.96 | 14 | COL1A1, NPPA, PNMT, ICAM1, UCP3, H19, FN1, PFKFB1, ASS1, CCL2, NR4A1, EGR2, HDAC9, LDLR | 4.73E-05 |
GO:0048545 | Response to steroid hormone | 11.61 | 14 | COL1A1, PNMT, ICAM1, UCP3, H19l, ELN, FN1, FBXO32, PFKFB1, ASS1, CCL2, FOSL1, SULT1A1, FOSB | 9.04E-06 |
GO:0051384 | Response to glucocorticoid | 13.78 | 13 | PNMT, ICAM1, UCP3, H19, ELN, FN1, FBXO32, PFKFB1, ASS1, CCL2, FOSL1, SULT1A1, FOSB | 1.03E-06 |
GO:0071229 | Cellular response to acid chemical | 12.49 | 12 | COL1A1, COL1A2, COL3A1, PDK4, ELN, FN1, ASS1, CCL2, CAMK2B, CCNB1, LDLR, ATP1A3 | 3.76E-06 |
GO:0030198 | Extracellular matrix organization | 12.63 | 11 | COL1A1, COL1A2, LOXL2, COL3A1, COL8A1, PTX3, ELN, FN1, FOXC2, SCARA3, HAS1 | 3.28E-06 |
GO:0043062 | Extracellular Structure organization | 11.26 | 11 | COL1A1, COL1A2, LOXL2, COL3A1, COL8A1, PTX3, ELN, FN1, FOXC2, SCARA3, HAS1 | 1.29E-05 |
GO:0031012 | Extracellular matrix | 9.42 | 11 | COL1A1, COL1A2, LOXL2, COL3A1, COL8A1, LRRC17, PTX3, ELN, FN1, SCARA3, THBS1 | 8.10E-05 |
GO:0005201 | Extracellular matrix structural constituent | 15.37 | 7 | COL1A1, COL1A2, COL3A1, COL8A1, ELN, FN1, SCARA3 | 2.11E-07 |
GO:0071549 | Cellular response to dexamethasone stimulus | 10.19 | 6 | PNMT, ICAM1, ELN, FBXO32, ASS1, CCL2 | 3.75E-05 |
GO:0071398 | Cellular response to fatty acid | 9.9 | 6 | PDK4, FN1, ASS1, CCL2, CCNB1, LDLR | 5.01E-05 |
GO:0044344 | Cellular response to fibroblast growth factor stimulus | 9.98 | 5 | COL1A1, ELN, CCL2, NR4A1, EGR3 | 4.64E-05 |
GO:0051310 | Metaphase plate congression | 9.59 | 5 | KIF22, GEM, CDCA8, CCNB1, CENPF | 6.82E-05 |
GO:0071774 | Response to fibroblast growth factor | 9.24 | 5 | COL1A1, ELN, CCL2, NR4A1, EGR3 | 9.72E-05 |
GO:0005584 | Collagen type I Trimer | 9.05 | 2 | COL1A1, COL1A2 | 1.17E-04 |
MR 40 wk | |||||
GO:0016020 | Membrane | 11.3 | 120 | LPIN1, ZMYND10, GPR22, PDE3A, PDE4D, HSPA13, ADCY1, KCND2, KCNK3, MPP5, MGRN1, ACOX1, PIP4K2B, CHRM2, OGDH, NPR3, MYOT, BMPR1A, ADRB1, PDPK1, SLC4A4, GSK3B, PHKB, CAVIN4, NCAM1, NCEH1, CLIC4, SC5D, SYNPO, CREB3L2, KIF1B, PI4K2A, MFAP3L, SEC23A, PRKAA2, PRKAB2, PRKACB, ZBTB16, BACE1, FAM210B, TGFA, SPTB, AKAP6, SLCO3A1, CORIN, CPD, MVB12B, CPNE3, HK2, ADIPOR2, MCU, NIPSNAP2, ANKH, YAP1, ANGPT1, ATF6, CAP2, RASL10B, ASS1, CBLC, XPR1, CDS1, CDS2, CDV3, MAN1A1, PRKAR2A, MTFR1, ZHX2, EXTL3, NAPEPLD, KPNA4, ZMPSTE24, SLC16A1, FOXO3, SLMAP, ENAH, LIN7C, ERC1, DNM1L, PRKCE, LIMS1, SLC38A1, MAGT1, CAMK2B, GJA3, GLG1, HCN2, HCN4, PDLIM5, PTGFR, PARD3B, GPAM, FZD1, B3GALT2, CHRNA1, PTPRN, PTPRT, CDH22, LMAN1, CSNK2A1, GNA13, CCND2, INSR, GNAO1, IRS2, ADRA1A, JPH1, JPH2, NDUFA1, LOC500331, ABCB7, GPD1L, PCYT1A, ABHD2, PCYOX1, MAOA, SLC25A30, LOC108348108, PCDH7, ATP1A3 | 1.24E-05 |
GO:0048513 | Animal organ development | 13.28 | 53 | LPIN1, ARHGAP5, PDE4D, COL11A2, ADCY1, NFIA, NFIB, KCNK3, QK, TGFBR3, BMPR1A, NTN1, ARID5B, CAVIN4, AMIGO1, NCAM1, CREB3L2, KIF5B, NCOA2, RPA1, ZBTB16, TBX5, SPTB, ESRRB, AK4, ADIPOR2, LBH, WFS1, ALPK3, RRM2B, YAP1, MICAL2, ANGPT1, ATF6, ASS1, PPARA, ZMPSTE24, TEAD1, FGF1, PROX1, HCN2, PDLIM5, HYAL1, TGFB2, SOX12, CDH22, CSNK2A1, CCND2, INSR, IRS2, ADRA1A, JPH1, LIFR | 1.71E-06 |
GO:0005886 | Plasma membrane | 10.85 | 76 | GPR22, PDE4D, HSPA13, ADCY1, KCND2, KCNK3, MPP5, MGRN1, ACOX1, PIP4K2B, CHRM2, MYOT, BMPR1A, ADRB1, PDPK1, GSK3B, PHKB, CAVIN4, NCAM1, CLIC4, PI4K2A, MFAP3L, PRKACB, ZBTB16, BACE1, TGFA, SPTB, AKAP6, SLCO3A1, CORIN, CPD, MVB12B, CPNE3, HK2, ADIPOR2, ANKH, ANGPT1, CAP2, CBLC, XPR1, CDV3, PRKAR2A, MTFR1, ZHX2, SLC16A1, SLMAP, ENAH, LIN7C, DNM1L, PRKCE, LIMS1, SLC38A1, MAGT1, GJA3, HCN2, HCN4, PTGFR, GPAM, FZD1, CHRNA1, PTPRN, PTPRT, CDH22, CSNK2A1, INSR, IRS2, ADRA1A, JPH1, JPH2, LOC500331, GPD1L, PCYT1A, ABHD2, PCYOX1, PCDH7, ATP1A3 | 1.93E-05 |
GO:0032501 | Multicellular organismal process | 10.63 | 71 | PDE3A, PDE4D, COL11A2, ADCY1, NFIB, NFIC, KCND2, TGFBR3, NPR3, BMPR1A, ADRB1, GSK3B, ARID5B, HIPK2, OPA3, BTBD9, NCAM1, CLIC4, REST, DDAH1, SYNPO, NCOA2, RPA1, PRKAA2, ZBTB16, TBX5, SOBP, ESRRB, CORIN, FTO, ADIPOR2, IGFBP5, WFS1, IF4EBP2, MN1, FBXO32, RRM2B, YAP1, ANGPT1, ATF6, RASL10B, PPARA, ZHX2, GFPT1, NAPEPLD, ZMPSTE24, TBX20, FOXO3, SLMAP, MTURN, XPNPEP3, TENT5C, DNM1L, LIMS1, SLC38A1, PROX1, MAGT1, CAMK2B, MAP1A, GJA3, HCN4, TGFB2, CHRNA1, MBNL1, GNA13, CCND2, INSR, ADRA1A, ADRA2C, JPH2, ATP1A3 | 2.42E-05 |
GO:0003008 | System process | 11.74 | 39 | PDE3A, PDE4D, COL11A2, ADCY1, NFIB, NFIC, KCND2, NPR3, ADRB1, OPA3, BTBD9, NCAM1, REST, DDAH1, SYNPO, SOBP, CORIN, WFS1, EIF4EBP2, FBXO32, RRM2B, YAP1, ANGPT1, ATF6, RASL10B, ZMPSTE24, TBX20, SLMAP, XPNPEP3, DNM1L, MAGT1, CAMK2B, MAP1A, GJA3, CHRNA1, CCND2, ADRA1A, ADRA2C, ATP1A3 | 7.94E-06 |
GO:0071495 | Cellular response to endogenous stimulus | 11.46 | 35 | LPIN1, PDE3A, PDE4D, BMPR1A, PDPK1, GSK3B, AMIGO1, REST, KIF1B, NCOA2, PRKAA2, BACE1, FAM210B, AKAP6, SESN3, CPEB4, IGFBP5, FBXO32, ASS1, GFPT1, ZMPSTE24, FOXO3, PRKCE, LIMS1, CAMK2B, HCN2, HCN4, HYAL1, PTGFR, GPAM, FZD1, CCND2, INSR, IRS2, ATP1A3 | 1.06E-05 |
GO:0045927 | Positive regulation of growth | 12.39 | 18 | TGFBR3, BMPR1A, ADRB1, NTN1, GSK3B, NCAM1, TBX5, AKAP6, WFS1, YAP1, EXTL3, TBX20, PROX1, HYAL1, GPAM, TGFB2, CSNK2A1, INSR | 4.15E-06 |
GO:0055024 | Regulation of cardiac muscle tissue development | 13.31 | 11 | TGFBR3, BMPR1A, ADRB1, GSK3B, NCAM1. TBX5, AKAP6, YAP1, PARA, TBX20, JPH2 | 1.65E-06 |
GO:0030165 | PDZ domain binding | 12.45 | 11 | ACOX1, TGFBR3, ADRB1, NCOA2, CRIM1, EXOC4, LIN7C, ERC1, HCN2, SNTB1, FZD1 | 3.91E-06 |
GO:0060420 | Regulation of heart growth | 12.92 | 10 | TGFBR3, BMPR1, AADRB1, NCAM1, TBX5, AKAP6, YAP1, PPARA, TBX20, PROX1 | 2.45E-06 |
GO:0019935 | Cyclic-nucleotide-mediated signaling | 10.8 | 11 | PDE3A, PDE4D, PDE7B, ADCY1, ADRB1, EIF4EBP2, PRKAR2A, PTGFR, GNA13, ADRA1A, ADRA2C | 2.03E-05 |
GO:0030018 | Z disc | 10.56 | 11 | NEBL, MYOT, SYNPO2, CAVIN4, SPHKAP, SYNPO, FBXO32, PDLIM5, ADRA1A, JPH1, JPH2 | 2.59E-05 |
GO:0019933 | cAMP-mediated signaling | 10.96 | 10 | PDE3A, PDE4D, PDE7B, ADCY1, ADRB1, EIF4EBP2, PTGFR, GNA13, ADRA1A, ADRA2C | 1.74E-05 |
GO:0046620 | Regulation of organ growth | 10.41 | 10 | TGFBR3, BMPR1A, ADRB1, NCAM1, TBX5, AKAP6, YAP1, PPARA, TBX20, PROX1 | 3.02E-05 |
GO:0060421 | Positive regulation of heart growth | 15.51 | 9 | TGFBR3, BMPR1A, ADRB1, NCAM1, TBX5, AKAP6, YAP1, TBX20, PROX1 | 1.84E-07 |
GO:0055025 | Positive regulation of cardiac muscle tissue development | 14.17 | 9 | TGFBR3, BMPR1A, ADRB1, GSK3B, NCAM1, TBX5, AKAP6, YAP1, TBX20 | 6.98E-07 |
GO:0046622 | Positive regulation of organ growth | 14 | 9 | TGFBR3, BMPR1A, ADRB1, NCAM1, TBX5, AKAP6, YAP1, TBX20, PROX1 | 8.30E-07 |
GO:0055021 | Regulation of cardiac muscle tissue growth | 11.45 | 9 | TGFBR3, BMPR1A, ADRB1, NCAM1, TBX5, AKAP6, YAP1, PPARA, TBX20 | 1.07E-05 |
GO:0045844 | Positive regulation of striated muscle tissue development | 11.21 | 9 | TGFBR3, BMPR1A, ADRB1, GSK3B, NCAM1, TBX5, AKAP6, YAP1, TBX20 | 1.35E-05 |
GO:0048636 | Positive regulation of muscle organ development | 11.21 | 9 | TGFBR3, BMPR1A, ADRB1, GSK3B, NCAM1, TBX5, AKAP6, YAP1, TBX20 | 1.35E-05 |
GO:1901863 | Positive regulation of muscle tissue development | 11.1 | 9 | TGFBR3, BMPR1A, ADRB1, GSK3B, NCAM1, TBX5, AKAP6, YAP1, TBX20 | 1.51E-05 |
GO:2000736 | Regulation of stem cell differentiation | 11.52 | 8 | BMPR1A, GSK3B, REST, TBX5, ESRRB, LBH, YAP1, TGFB2 | 9.95E-06 |
GO:0055023 | Positive regulation of cardiac muscle tissue growth | 13.54 | 8 | TGFBR3, BMPR1A, ADRB1, NCAM1, TBX5, AKAP6, YAP1, TBX20 | 1.32E-06 |
GO:0060045 | Positive regulation of cardiac muscle cell proliferation | 11.47 | 6 | TGFBR3, BMPR1A, NCAM1, TBX5, YAP1, TBX20 | 1.04E-05 |
GO:1902459 | Positive regulation of stem cell population maintenance | 15.58 | 5 | REST, ESRRB, LBH, YAP1, TEAD1 | 1.71E-07 |
GO, Gene Ontology; MR, mitral regurgitation, BP, Biological Process.
TABLE E8.
Three PANTHER-classified pathways of the up- and down-regulated genes after MR onset
MR 2-wk up-regulated genes | MR 2-wk down-regulated genes |
---|---|
p53 pathway | p53 pathway |
Thyrotropin-releasing hormone receptor signaling pathway | Endothelin signaling pathway |
DNA replication | Heterotrimeric G-protein signaling pathway-Gi alpha– and Gs alpha–mediated pathway |
MR 10-wk up-regulated genes | MR 10-wk down-regulated genes |
Angiotensin II-stimulated signaling through G proteins and beta-arrestin | Integrin signaling pathway |
Oxidative stress response | DNA replication |
Serine glycine biosynthesis | Nicotinic acetylcholine receptor signaling pathway |
MR 20-wk up-regulated genes | MR 20-wk down-regulated genes |
TGF-beta signaling pathway | p53 pathway |
Oxidative stress response | Inflammation mediated by cytokine and chemokine signaling pathway |
Adrenaline and noradrenaline biosynthesis | Integrin signaling pathway |
MR 40-wk up-regulated genes | MR 40-wk down-regulated genes |
Heterotrimeric G-protein signaling pathway–Gi alpha and Gs alpha–mediated pathway | EGF receptor signaling pathway |
Dopamine receptor mediated signaling pathway | Integrin signaling pathway |
Adrenaline and noradrenaline biosynthesis | Cell cycle |
MR, Mitral regurgitation; TGF, transforming growth factor; EGF, epidermal growth factor.
TABLE E9.
KEGG analysis of the significantly altered pathways, comparing MR versus sham differentially expressed genes
Category | KEGG pathway | Enrichment score | Gene count | Gene symbol | P value |
---|---|---|---|---|---|
MR 2 wk | |||||
path:rno04110 | Cell cycle | 13.17 | 9 | BUB1B, CCNA2, CCNB1, CCNB2, CDC20, CDC6, CDK1, E2F1, ESPL1 | 1.91E-06 |
path:rno04218 | Cellular senescence | 10.22 | 9 | CCNA2, CCNB1, CCNB2, CDK1, E2F1, FOXM1, IGFBP3, IL6, MYBL2 | 3.65E-05 |
path:rno04115 | p53 signaling pathway | 7.49 | 5 | CCNB1, CCNB2, CDK1, IGFBP3, RRM2 | 5.61E-04 |
path:rno04914 | Progesterone-mediated oocyte maturation | 6.78 | 5 | CCNA2, CCNB1, CCNB2, CDK1, KIF22 | 1.14E-03 |
path:rno05166 | Human T-cell leukemia virus 1 infection | 6.59 | 8 | BUB1B, CCNA2, CCNB2, CDC20, E2F1, ESPL1, IL6, MSX2 | 1.37E-03 |
path:rno04614 | Renin-angiotensin system | 6.55 | 3 | CMA1, CPA3, MCPT1L1 | 1.43E-03 |
path:rno04061 | Viral protein interaction with cytokine and cytokine receptor | 5.71 | 4 | CCL7, CXCL14, IL6, PF4 | 3.33E-03 |
path:rno04114 | Oocyte meiosis | 5.62 | 5 | CCNB1, CCNB2, CDC20, CDK1, ESPL1 | 3.61E-03 |
path:rno04060 | Cytokine-cytokine receptor interaction | 4.97 | 6 | CCL7, CXCL14, EPOR, IL6, LTB, PF4 | 6.92E-03 |
path:rno04657 | IL-17 signaling pathway | 3.48 | 3 | CCL7, FOSB, IL6 | .03 |
path:rno05020 | Prion diseases | 3.26 | 2 | C6, IL6 | .04 |
MR 10 wk | |||||
path:rno04925 | Aldosterone synthesis and secretion | 6.54 | 5 | AGT, ATP1A3, CAMK2B, LDLR, NPPA | 1.45E-03 |
path:rno00350 | Tyrosine metabolism | 6.37 | 3 | HPD, MAOA, PNMT | 1.70E-03 |
path:rno04927 | Cortisol synthesis and secretion | 6.25 | 4 | AGT, KCNA4, KCNK2, LDLR | 1.93E-03 |
path:rno00360 | Phenylalanine metabolism | 5.17 | 2 | HPD, MAOA | 5.67E-03 |
path:rno04080 | Neuroactive ligand-receptor interaction | 5.17 | 6 | AGT, CHRNA1, CNR2, MC4R, NPFF, SSTR3 | 5.70E-03 |
path:rno04934 | Cushing syndrome | 4.65 | 5 | AGT, CAMK2B, KCNA4, KCNK2, LDLR | 9.53E-03 |
path:rno00982 | Drug metabolism - cytochrome P450 | 4.52 | 3 | FMO2, MAOA, UGT1A1 | .01 |
path:rno00340 | Histidine metabolism | 4.43 | 2 | ASPA, MAOA | .01 |
path:rno05031 | Amphetamine addiction | 4.02 | 3 | CAMK2B, FOSB, MAOA | .02 |
path:rno04971 | Gastric acid secretion | 3.82 | 3 | ATP1A3, CAMK2B, KCNK2 | .02 |
path:rno04974 | Protein digestion and absorption | 3.65 | 3 | ATP1A3, COL17A1, COL1A1 | .03 |
path:rno04911 | Insulin secretion | 3.44 | 3 | ATP1A3, CAMK2B, KCNN3 | .03 |
path:rno00250 | Alanine, aspartate, and glutamate metabolism | 3.36 | 2 | AGT, ASPA | .03 |
path:rno00140 | Steroid hormone biosynthesis | 3.06 | 2 | HSD17B1, UGT1A1 | .05 |
path:rno00260 | Glycine, serine, and threonine metabolism | 3.06 | 2 | AGT, MAOA | .05 |
path:rno04913 | Ovarian steroidogenesis | 3.06 | 2 | HSD17B1, LDLR | .05 |
path:rno05030 | Cocaine addiction | 2.9 | 2 | FOSB, MAOA | .05 |
MR 20 wk | |||||
path:rno04974 | Protein digestion and absorption | 12.4 | 7 | ATP1A3, COL1A1, COL1A2, COL3A1, ELN, PRCP, SLC1A5 | 4.11E-06 |
path:rno04933 | AGE-RAGE signaling pathway in diabetic complications | 9.29 | 7 | CCL2, COL1A1, COL1A2, COL3A1, FN1, ICAM1, VCAM1 | 9.22E-05 |
path:rno05144 | Malaria | 6.37 | 4 | CCL2, ICAM1, THBS1, VCAM1 | 1.71E-03 |
path:rno04925 | Aldosterone synthesis and secretion | 6.22 | 5 | ATP1A3, CAMK2B, LDLR, NPPA, NR4A1 | 1.99E-03 |
path:rno00350 | Tyrosine metabolism | 6.17 | 3 | HPD, MAOA, PNMT | 2.09E-03 |
path:rno05143 | African trypanosomiasis | 5.4 | 3 | ICAM1, NPPA, VCAM1 | 4.54E-03 |
path:rno04512 | ECM-receptor interaction | 5.11 | 4 | COL1A1, COL1A2, FN1, THBS1 | 6.06E-03 |
path:rno00360 | Phenylalanine metabolism | 5.03 | 2 | HPD, MAOA | 6.52E-03 |
path:rno05146 | Amoebiasis | 4.56 | 4 | COL1A1, COL1A2, COL3A1, FN1 | .01 |
path:rno04934 | Cushing syndrome | 4.39 | 5 | CAMK2B, LDLR, NR4A1, RASD1, WNT9B | .01 |
path:rno05031 | Amphetamine addiction | 3.83 | 3 | CAMK2B, FOSB, MAOA | .02 |
path:rno05034 | Alcoholism | 3.64 | 4 | FOSB, HDAC9, MAOA, NPY | .03 |
path:rno04657 | IL-17 signaling pathway | 3.3 | 3 | CCL2, FOSB, FOSL1 | .04 |
path:rno04115 | p53 signaling pathway | 3.26 | 3 | CCNB1, CCNB2, THBS1 | .04 |
path:rno05418 | Fluid shear stress and atherosclerosis | 2.92 | 4 | ASS1, CCL2, ICAM1, VCAM1 | .05 |
MR 40 wk | |||||
path:rno04932 | Nonalcoholic fatty liver disease (NAFLD) | 14.21 | 36 | ADIPOR2, AKT2, BAX, COX4I1, COX4I2, COX6B1, COX6C, COX7A2, COX7C, COX8B, ERN1, GSK3B, INSR, IRS1, IRS2, ITCH, NDUFA1, NDUFA10L1, NDUFA11, NDUFA6, NDUFA7, NDUFB2, NDUFB3, NDUFB7, NDUFB9, NDUFS1, NDUFS5, NDUFS6, PIK3R1, PPARA, PRKAA2, PRKAB2, RXRA, UQCR10, UQCR11, UQCRB | 6.76E-07 |
path:rno03010 | Ribosome | 13.9 | 35 | FAU, MRPL14, MRPL27, RPL11, RPL13A, RPL18, RPL21, RPL22, RPL23A, RPL27, RPL27A, RPL30, RPL31, RPL32, RPL35A, RPL36, RPL36AL, RPL37A, RPL38, RPL39, RPL41, RPLP1, RPLP2, RPS10, RPS12, RPS15, RPS15A, RPS17, RPS18, RPS19, RPS24, RPS27A, RPS28, RPS8, RPS9 | 9.17E-07 |
path:rno04714 | Thermogenesis | 13.04 | 44 | ADCY1, ADCY5, ATF2, ATP5E, ATP5G3, ATP5H, ATP5I, ATP5J2, ATP5L, COA5, COX17, COX4I1, COX4I2, COX6B1, COX6C, COX7A2, COX7C, COX8B, CREB3L2, MAPK11, NDUFA1, NDUFA10L1, NDUFA11, NDUFA6, NDUFA7, NDUFB2,. NDUFB3, NDUFB7, NDUFB9, NDUFS1, NDUFS5, NDUFS6, PPARGC1A, PRKAA2, PRKAB2, PRKACB, PRKG1, RPS6KB2, SMARCC1, TSC1, UQCR10, UQCR11, UQCRB, ZFP516 | 2.18E-06 |
path:rno00190 | Oxidative phosphorylation | 13.01 | 30 | ATP5E, ATP5G3, ATP5H, ATP5I, ATP5J2, ATP5L, ATP6V0B, COX17, COX4I1, COX4I2, COX6B1, COX6C, COX7A2, COX7C, COX8B, NDUFA1, NDUFA10L1, NDUFA11, NDUFA6, NDUFA7, NDUFB2, NDUFB3, NDUFB7, NDUFB9, NDUFS1, NDUFS5, NDUFS6, UQCR10, UQCR11, UQCRB | 2.23E-06 |
path:rno05016 | Huntington disease | 11.36 | 36 | AP2S1, ATP5E, ATP5G3, ATP5H, BAX, BBC3, COX4I1, COX4I2, COX6B1, COX6C, COX7A2, COX7C, COX8B, CREB3L2, DCTN4, GNAQ, NDUFA1, NDUFA10L1, NDUFA11, NDUFA6, NDUFA7, NDUFB2, NDUFB3, NDUFB7, NDUFB9, NDUFS1, NDUFS5, NDUFS6, POLR2H, POLR2I, POLR2L, PPARGC1A, REST, UQCR10, UQCR11, UQCRB | 1.16E-05 |
path:rno05010 | Alzheimer disease | 10.43 | 35 | APH1B, ATF6, ATP5E, ATP5G3, ATP5H, BACE1, COX4I1, E60COX4I2, COX6B1, COX6C, COX7A2, COX7C, COX8B, ERN1, GNAQ, GSK3B, IDE, LPL, NDUFA1, NDUFA10L1, NDUFA11, NDUFA6, NDUFA7, NDUFB2, NDUFB3, NDUFB7, NDUFB9, NDUFS1, NDUFS5, NDUFS6, NOS1, PPP3CB, UQCR10, UQCR11, UQCRB | 2.94E-05 |
path:rno05012 | Parkinson disease | 9.07 | 28 | ADCY5, ATP5E, ATP5G3, ATP5H, COX4I1, COX4I2, COX6B1, COX6C, COX7A2, COX7C, COX8B, NDUFA1, NDUFA10L1, NDUFA11, NDUFA6, NDUFA7, NDUFB2, NDUFB3, NDUFB7, NDUFB9, NDUFS1, NDUFS5, NDUFS6, PRKACB, PRKN, UQCR10, UQCR11, UQCRB | 1.15E-04 |
path:rno04211 | Longevity regulating pathway | 7.98 | 20 | ADCY1, ADCY5, ADIPOR2, AKT2, ATF2, BAX, CREB3L2, FOXO1, FOXO3, INSR, IRS1, IRS2, PIK3R1, PPARGC1A, PRKAA2, PRKAB2, PRKACB, RPS6KB2, SESN3, TSC1 | 3.43E-04 |
path:rno00564 | Glycerophospholipid metabolism | 7.98 | 20 | AGPAT4, CDS1, CDS2, CHPT1, DGKG, DGKQ. DGKZ, GPAM, GPAT3, GPD1, GPD1L, GPD2, LCAT, LPCAT2, LPGAT1, LPIN1, PCYT1A, PLA2G2A, PLA2G4B, PLD4 | 3.43E-04 |
path:rno04723 | Retrograde endocannabinoid signaling | 7.54 | 25 | ADCY1, ADCY5, GNAO1, GNAQ, GNG11, GNG7, GNGT2, KCNJ3, MAPK11, NAPEPLD, NDUFA1, NDUFA10L1, NDUFA11, NDUFA6, NDUFA7, NDUFB2, NDUFB3, NDUFB7, NDUFB9, NDUFS1, NDUFS5, NDUFS6, PRKACB, PRKCG, SLC17A7 | 5.30E-04 |
path:rno04922 | Glucagon signaling pathway | 7.24 | 21 | ACACA, AKT2, ATF2, CDK1, CAMK2B, CAMK2G, CREB3L2, FBP2, FOXO1, GCGR, GNAQ, PCK1, PCK2, PHKA1, PHKB, PPARA, PPARGC1A, PPP3CB, PRKAA2, PRKAB2, PRKACB | 7.16E-04 |
path:rno04910 | Insulin signaling pathway | 6.88 | 26 | ACACA, AKT2, CRK, CRKL, FBP2, FOXO1, GSK3B, HK2, INSR, IRS1, IRS2, PCK1, PCK2, PDPK1, PHKA1, PHKB, PIK3R1, PPARGC1A, PPP1R3A, PRKAA2, PRKAB2, PRKACB, PRKAR2A, RPS6KB2, SH2B2, TSC1 | 1.03E-03 |
path:rno05032 | Morphine addiction | 6.87 | 17 | ADCY1, ADCY5, ADORA1, ARRB1, GNAO1, GNG11, GNG7, GNGT2, KCNJ3, PDE1C, PDE3A, PDE4C, PDE4D, PDE7A, PDE7B, PRKACB, PRKCG | 1.04E-03 |
path:rno04213 | Longevity regulating pathway—multiple species | 6.63 | 15 | ADCY1, ADCY5, AKT2, EIF4EBP2, FOXO1, FOXO3, HSPA1B, INSR, IRS1, IRS2, PIK3R1, PRKAA2, PRKAB2, PRKACB, RPS6KB2 | 1.32E-03 |
path:rno04152 | AMPK signaling pathway | 6.54 | 24 | ACACA, ADIPOR2, ADRA1A, AKT2, CREB3L2, FBP2, FOXO1, FOXO3, INSR, IRS1, IRS2, PCK1, PCK2, PDPK1, PFKFB4, PIK3R1, PPARGC1A, PPP2R3A, PPP2R5C, PRKAA2, PRKAB2, RAB14, RPS6KB2, TSC1 | 1.44E-03 |
path:rno04923 | Regulation of lipolysis in adipocytes | 6.43 | 13 | ABHD5, ADCY1, ADCY5, ADORA1, ADRB1, AKT2, FABP4, INSR, IRS1, IRS2, PIK3R1, PRKACB, PRKG1 | 1.61E-03 |
path:rno04713 | Circadian entrainment | 5.42 | 18 | ADCY1, ADCY5, CAMK2B, CAMK2G, GNAO1, GNAQ, GNG11, GNG7, GNGT2, KCNJ3, NOS1, NOS1AP, PER3, PRKACB, PRKCG, PRKG1, RASD1, RYR2 | 4.44E-03 |
path:rno04725 | Cholinergic synapse | 5.19 | 19 | ADCY1, ADCY5, AKT2, BCL2, CAMK2B, CAMK2G, CHRM2, CREB3L2, GNAO1, GNAQ, GNG11, GNG7, GNGT2, KCNJ12, KCNJ2, KCNJ3, PIK3R1, PRKACB, PRKCG | 5.59E-03 |
path:rno00230 | Purine metabolism | 5.04 | 21 | ADCY1, ADCY5, AK4, AK6, AMPD1, AMPD3, ENTPD5, GDA, NME1, NME2, NME3, NT5C, NT5C1A, PDE1C, PDE3A, PDE4C, PDE4D, PDE7A, PDE7B, RRM2, RRM2B | 6.45E-03 |
path:rno04260 | Cardiac muscle contraction | 4.64 | 15 | ATP1A3, CACNA2D1, COX4I1, COX4I2, COX6B1, COX6C, COX7A2, COX7C, COX8B, MYL4, RYR2, TNNI3, UQCR10, UQCR11, UQCRB | 9.65E-03 |
path:rno04931 | Insulin resistance | 4.53 | 20 | AKT2, CREB3L2, FOXO1, GFPT1, GSK3B, INSR, IRS1, IRS2, MGEA5, PCK1, PCK2, PDPK1, PIK3R1, PPARA, PPARGC1A, PPP1R3A, PRKAA2, PRKAB2, PRKCE, RPS6KB2 | .01 |
path:rno04911 | Insulin secretion | 4.47 | 14 | ADCY1, ADCY5, ATF2, ATP1A3, CAMK2B, CAMK2G, CREB3L2, GNAQ, KCNMB4, KCNN4, PRKACB, PRKCG, RYR2, STX1A | .01 |
path:rno04022 | cGMP-PKG signaling pathway | 4.4 | 26 | ADCY1, ADCY5, ADORA1, ADRA1A, ADRA2B, ADRA2C, ADRB1, AKT2, ATF2, ATP1A3, CREB3L2, GATA4, GNA13, GNAQ, INSR, IRS1, IRS2, KCNMB4, MEF2A, MEF2D, MYL9, MYLK3, PDE3A, PPP3CB, PRKCE, PRKG1 | .01 |
path:rno04068 | FoxO signaling pathway | 4.36 | 22 | AKT2, CCNB2, CCND2, EGF, FBXO32, FOXO1, FOXO3, HOMER1, INSR, IRS1, IRS2, MAPK11, PCK1, PCK2, PDPK1, PIK3R1, PLK1, PRKAA2, PRKAB2, SETD7, TGFB2, TGFBR2 | .01 |
path:rno00240 | Pyrimidine metabolism | 4.28 | 11 | DCTD, ENTPD5, NME1, NME2, NME3, NT5C, NT5C1A, RRM2, RRM2B, TK1, UCK2 | .01 |
path:rno04928 | Parathyroid hormone synthesis, secretion, and action | 4.23 | 17 | ADCY1, ADCY5, ARRB1, ATF2, BCL2, CREB3L2, GNA13, GNAQ, LRP6, MEF2A, MEF2D, PDE4C, PDE4D, PRKACB, PRKCG, RXRA, VDR | .01 |
path:rno04914 | Progesterone-mediated oocyte maturation | 4.17 | 16 | ADCY1, ADCY5, AKT2, ANAPC11, CCNB2, CDC27, CDK1, CPEB1, CPEB4, KIF22, MAD2L2, MAPK11, PIK3R1, PKMYT1, PLK1, PRKACB | .02 |
path:rno04728 | Dopaminergic synapse | 4.1 | 21 | ADCY5, AKT2, ATF2, CAMK2B, CAMK2G, CREB3L2, GNAO1, GNAQ, GNG11, GNG7, GNGT2, GSK3B, KCNJ3, KIF5B, MAOA, MAPK11, PPP2R3A, PPP2R5C, PPP3CB, PRKACB, PRKCG | .02 |
path:rno04724 | Glutamatergic synapse | 4.01 | 17 | ADCY1, ADCY5, GNAO1, GNAQ, GNG11, GNG7, GNGT2, GRIK4, HOMER1, KCNJ3, PLA2G4B, PPP3CB, PRKACB, PRKCG, SHANK1, SLC17A7, SLC38A1 | .02 |
path:rno04114 | Oocyte meiosis | 3.94 | 19 | ADCY1, ADCY5, ANAPC11, CAMK2B, CAMK2G, CCNB2, CDC20, CDC27, CDK1, CPEB1, CPEB4, MAD2L2, MAPK11, PKMYT1, PLK1, PPP2R5C, PPP3CB, PRKACB, SLK | .02 |
path:rno04926 | Relaxin signaling pathway | 3.92 | 20 | ADCY1, ADCY5, AKT2, ARRB1, ATF2, COL1A1, COL3A1, CREB3L2, GNA15, GNAO1, GNG11, GNG7, GNGT2, MAP2K4, MAPK11, NOS1, PIK3R1, PRKACB, RXFP1, TGFBR2 | .02 |
path:rno00512 | Mucin type O-glycan biosynthesis | 3.82 | 6 | C1GALT1, GALNT1, GALNT10, GALNT17, GALNT6, ST3GAL1 | .02 |
path:rno04978 | Mineral absorption | 3.8 | 9 | ATOX1, ATP1A3, ATP7A, MT2A, SLC26A6, SLC26A9, SLC9A3, STEAP2, VDR | .02 |
path:rno05231 | Choline metabolism in cancer | 3.8 | 17 | AKT2, CHPT1, DGKG, DGKQ, DGKZ, EGF, PCYT1A, PDGFA, PDGFD, PDPK1, PIK3R1, PLA2G4B, PLCG1, PRKCG, RPS6KB2, SLC22A1, TSC1 | .02 |
path:rno04916 | Melanogenesis | 3.77 | 15 | ADCY1, ADCY5, CAMK2B, CAMK2G, CREB3L2, FZD1, FZD5, FZD8, GNAO1, GNAQ, GSK3B, POMC, PRKACB, PRKCG, WNT9B | .02 |
path:rno04012 | ErbB signaling pathway | 3.55 | 15 | AKT2, CAMK2B, CAMK2G, CRK, CRKL, EGF, GSK3B, MAP2K4, PAK2, PAK6, PIK3R1, PLCG1, PRKCG, RPS6KB2, TGFA | .03 |
path:rno00100 | Steroid biosynthesis | 3.5 | 5 | DHCR24, EBP, HSD17B7, SC5D, SOAT1 | .03 |
path:rno04920 | Adipocytokine signaling pathway | 3.41 | 13 | ADIPOR2, AKT2, IRS1, IRS2, PCK1, PCK2, POMC, PPARA, PPARGC1A, PRKAA2, PRKAB2, RXRA, TRADD | .03 |
path:rno04371 | Apelin signaling pathway | 3.39 | 21 | ADCY1, ADCY5, AKT2, APLNR, GNA13, GNAQ, GNG11, GNG7, GNGT2, MEF2A, MEF2D, MYL4, MYLK3, NOS1, PPARGC1A, PRKAA2, PRKAB2, PRKACB, PRKCE, RPS6KB2, RYR2 | .03 |
path:rno04020 | Calcium signaling pathway | 3.06 | 24 | ADCY1, ADRA1A, ADRB1, CAMK2B, CAMK2G, CHRM2, GNA15, GNAQ, MCU, MYLK3, NOS1, NTSR1, PDE1C, PHKA1, PHKB, PLCE1, PLCG1, PPP3CB, PRKACB, PRKCG, PTGFR, RYR2, TBXA2R, TPCN2 | .05 |
path:rno04971 | Gastric acid secretion | 3.04 | 11 | ADCY1, ADCY5, ATP1A3, CAMK2B, CAMK2G, GNAQ, KCNE2, KCNJ2, MYLK3, PRKACB, PRKCG | .05 |
path:rno00531 | Glycosaminoglycan degradation | 2.91 | 4 | ARSB, GNS, HYAL1, IDUA | .05 |
KEGG, Kyoto Encyclopedia of Genes and Genomes; ECM, extracellular matrix; IL, interleukin.
Footnotes
Webcast
You can watch a Webcast of this AATS meeting presentation by going to: https://aats.blob.core.windows.net/media/20AM/Presentations/Invasive%20Hemodynamics%20and%20Transcript.mp4.
Conflict of Interest Statement
M.P. received consulting fees from Heart Repair Technologies, Inc, which did not have any role in this study. All other authors reported no conflicts of interest.
The Journal policy requires editors and reviewers to disclose conflicts of interest and to decline handling or reviewing manuscripts for which they may have a conflict of interest. The editors and reviewers of this article have no conflicts of interest.
References
- 1.Nkomo VT, Gardin JM, Skelton TN, Gottdiener JS, Scott CG, Enriquez-Sarano M. Burden of valvular heart diseases: a population-based study. Lancet. 2006;368:1005–11. [DOI] [PubMed] [Google Scholar]
- 2.Apostolidou E, Maslow AD, Poppas A. Primary mitral valve regurgitation: update and review. Glob Cardiol Sci Pract. 2017;3:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Levine RA, Hagege AA, Judge DP, Padala M, Dal-Bianco JP, Aikawa E, et al. Mitral valve disease—morphology and mechanisms. Nat Rev Cardiol. 2015; 12:689–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Braunwald E, Welch GH, Sarnoff SJ. Hemodynamic effects of quantitativelyvaried experimental mitral regurgitation. Circ Res. 1957;5:539–45. [DOI] [PubMed] [Google Scholar]
- 5.Carabello BA, Crawford FA. Valvular heart disease. N Engl J Med. 1997;337: 32–41. [DOI] [PubMed] [Google Scholar]
- 6.Nishimura RA, Otto CM, Bonow RO, Carabello BA, Erwin JP III, Fleisher LA,et al. 2017 AHA/ACC focused update of the 2014 AHA/ACC guideline for the management of patients with valvular heart disease. Circulation. 2017;135: 1159–95. [DOI] [PubMed] [Google Scholar]
- 7.Gaasch WH, Meyer TE. Left ventricular response to mitral regurgitation implications for management. Circulation. 2008;118:2298–303. [DOI] [PubMed] [Google Scholar]
- 8.Kang DH, Kim JH, Rim JH, Kim MJ, Yun SC, Song JM, et al. Comparison of early surgery versus conventional treatment in asymptomatic severe mitral regurgitation. Circulation. 2009;119:797–804. [DOI] [PubMed] [Google Scholar]
- 9.Pu M, Gao Z, Li J, Sinoway L, Davidson WR. Development of a new animalmodel of chronic mitral regurgitation in rats under transesophageal echocardiographic guidance. J Am Soc Echocardiogr. 2005;18:468–74. [DOI] [PubMed] [Google Scholar]
- 10.Corporan D, Onohara D, Hernandez-Merlo R, Sielicka A, Padala M. Temporal changes in myocardial collagen, matrix metalloproteinases, and their tissue inhibitors in the left ventricular myocardium in experimental chronic mitral regurgitation in rodents. Am J Physiol Heart Circ Physiol. 2018;315: H1269–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Onohara D, Corporan D, Hernandez-Merlo R, Guyton RA, Padala M. Mitralregurgitation worsens cardiac remodeling in ischemic cardiomyopathy in an experimental model. J Thorac Cardiovasc Surg. 2020;160:e107–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Sengupta P The laboratory rat: relating its age with human’s. Int J Prev Med. 2013;4:624–30. [PMC free article] [PubMed] [Google Scholar]
- 13.Zoghbi WA, Enriquez-Sarano M, Foster E, Grayburn PA, Kraft CD, Levine RA,et al. Recommendations for evaluation of the severity of native valvular regurgitation with two-dimensional and Doppler echocardiography. J Am Soc Echocardiogr. 2003;16:777–802. [DOI] [PubMed] [Google Scholar]
- 14.Liu Z, Hilbelink DR, Crockett WB, Gerdes AM. Regional changes in hemodynamics and cardiac myocyte size in rats with aortocaval fistulas. Circ Res. 1991;69:52–8. [DOI] [PubMed] [Google Scholar]
- 15.Thomas DP, Phillips SJ, Bove AA. Myocardial morphology and blood flow distribution in chronic volume-overload hypertrophy in dogs. Basic Res Cardiol. 1984;79:379–88. [DOI] [PubMed] [Google Scholar]
- 16.Ryan TD, Rothstein EC, Aban I, Tallaj JA, Husain A, Lucchesi PA, et al. Leftventricular eccentric remodeling and matrix loss are mediated by bradykinin and precede cardiomyocyte elongation in rats with volume overload. J Am Coll Cardiol. 2007;49:811–21. [DOI] [PubMed] [Google Scholar]
- 17.Zheng J, Chen Y, Pat B, Dell’italia LA, Tillson M, Dillon AR, et al. Microarrayidentifies extensive downregulation of noncollagen extracellular matrix and profibrotic growth factor genes in chronic isolated mitral regurgitation in the dog. Circulation. 2009;119:2086–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Chen Y-W, Pat B, Gladden JD, Zheng J, Powell P, Wei CC, et al. Dynamic molecular and histopathological changes in the extracellular matrix and inflammation in the transition to heart failure in isolated volume overload. Am J Physiol Heart Circ Physiol. 2011;300:H2251–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kim KH, Kim HM, Park JS, Kim YJ. Differential transcriptome profile and exercise capacity in cardiac remodeling by pressure overload versus volume overload. J Cardiovasc Imaging. 2019;27:50–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rodriguez F, Langer F, Harrington KB, Tibayan FA, Zasio MK, Cheng A, et al. Importance of mitral valve second-order chordae for left ventricular geometry, wall thickening mechanics, and global systolic function. Circulation. 2004; 110(supp II):II115–22. [DOI] [PubMed] [Google Scholar]
- 21.Schuman ML, Landa MS, Toblli JE, Peres Diaz LS, Alvarez AL, Finkielman S,et al. Cardiac thyrotropin-releasing hormone mediates left ventricular hypertrophy in spontaneously hypertensive rats. Hypertension. 2011;57:103–9. [DOI] [PubMed] [Google Scholar]
- 22.Mak TW, Hauck L, Grothe D, Billia F. P53 regulates the cardiac transcriptome.Proc Natl Acad Sci U S A. 2017;114:2331–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Nomura S, Satoh M, Fujita T, Higo T, Sumida T, Ko T, et al. Cardiomyocyte geneprograms encoding morphological and functional signatures in cardiac hypertrophy and failure. Nat Commun. 2018;9:4435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kim KH, Rosen A, Bruneau BG, Hui CC, Backx PH. Iroquois homeodomaintranscription factors in heart development and function. Circ Res. 2012;110: 1513–24. [DOI] [PubMed] [Google Scholar]
- 25.Fiorillo C, Nediani C, Ponziani V, Giannini L, Celli A, Nassi N, et al. Cardiacvolume overload rapidly induces oxidative stress-mediated myocyte apoptosis and hypertrophy. Biochim Biophys Acta. 2005;1741:173–82. [DOI] [PubMed] [Google Scholar]
- 26.Yancey DM, Guichard JL, Ahmed MI, Zhou L, Murphy MP, Johnson MS, et al. Cardiomyocyte mitochondrial oxidative stress and cytoskeletal breakdown in the heart with a primary volume overload. Am J Physiol Heart Circ Physiol. 2015; 308:H651–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Robison P, Caporizzo MA, Ahmadzadeh H, Robison P, Caporizzo MA,Ahmadzadeh H, et al. Detyrosinated microtubules buckle and bear load in contracting cardiomyocytes et al. Detyrosinated microtubules buckle and bear load in contracting cardiomyocytes. Science. 2016;352:aaf0659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zablocki D, Sadoshima J. Angiotensin II and oxidative stress in the failing heart.Antioxid Redox Signal. 2013;19:1095–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Gladden JD, Ahmed MI, Litovsky SH, Schiros CG, Lloyd SG, Gupta H, et al. Oxidative stress and myocardial remodeling in chronic mitral regurgitation. Am J Med Sci. 2011;342:114–9. [DOI] [PubMed] [Google Scholar]
- 30.Brancaccio M, Hirsch E, Notte A, Selvetella G, Lembo G, Tarone G. Integrin signalling: the tug-of-war in heart hypertrophy. Cardiovasc Res. 2006;70:422–33. [DOI] [PubMed] [Google Scholar]
- 31.Zeng S, Liu A, Dai L, Yu X, Zhang Z, Xiong Q, et al. Prognostic value of TOP2Ain bladder urothelial carcinoma and potential molecular mechanisms. BMC Cancer. 2019;19:604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dobaczewski M, Chen W, Frangogiannis NG. Transforming growth factor(TGF)-b signaling in cardiac remodeling introduction: the biology of TGF-b. J Mol Cell Cardiol. 2011;51:600–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Cavallotti C, Mancone M, Bruzzone P, Sabbatini M, Mignini F. Dopamine receptor subtypes in the native human heart. Heart Vessels. 2010;25:432–7. [DOI] [PubMed] [Google Scholar]
- 34.Chen Q, Thompson J, Hu Y, Das A, Lesnefsky EJ. Cardiac specific knockout ofp53 decreases ER stress-induced mitochondrial damage. Front Cardiovasc Med. 2019;6:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kaye DM, Lambert GW, Lefkovits J, Morris M, Jennings G, Esler MD. Neurochemical evidence of cardiac sympathetic activation and increased central nervous system norepinephrine turnover in severe congestive heart failure. J Am Coll Cardiol. 1994;23:570–8. [DOI] [PubMed] [Google Scholar]
- 36.Liu L, An X, Li Z, Song Y, Li L, Zuo S, et al. The H19 long noncoding RNA is anovel negative regulator of cardiomyocyte hypertrophy. Cardiovasc Res. 2016; 111:56–65. [DOI] [PubMed] [Google Scholar]
- 37.Chen CY, Caporizzo MA, Bedi K, Vite A, Bogush AI, Robison P, et al. Suppression of detyrosinated microtubules improves cardiomyocyte function in human heart failure. Nat Med. 2018;24:1125–233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Thomson MJ, Frenneaux MP, Kaski JC. Antioxidant treatment for heart failure: friend or foe? Q J Med. 2009;102:305–10. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
VIDEO 1. Video showing the operative procedure of creating mitral regurgitation on the beating heart in the rat, using echocardiographic guidance. Video available at: https://www.jtcvs.org/article/S0022-5223(20)32784-7/fulltext.
VIDEO 2. Video depicting the beating heart in a rat from the sham group, depicting a smaller left atrium and left ventricle. Video available at: https://www.jtcvs.org/article/S0022-5223(20)32784-7/fulltext.
VIDEO 3. Video depicting the beating heart in a rat after 40 weeks of mitral regurgitation, depicting a severely enlarged left atrium and left ventricle. Video available at: https://www.jtcvs.org/article/S0022-5223(20)32784-7/fulltext.