Skip to main content

Abstract

Background:

The sino atrial node (SAN) is characterized by the microenvironment of pacemaker cardiomyocytes (PCs) encased with fibroblasts. An altered microenvironment leads to rhythm failure. Operable cell or tissue models are either generally lacking or difficult to handle. The biological process behind the milieu of SANs to evoke pacemaker rhythm is unknown. We explored how fibroblasts interact with PCs and regulate metabolic reprogramming and rhythmic activity in the SAN.

Methods:

Tbx18 (T-box transcription factor 18)-induced PCs and fibroblasts were used for cocultures and engineered tissues, which were used as the in vitro models to explore how fibroblasts regulate the functional integrity of SANs. RNA-sequencing, metabolomics, and cellular and molecular techniques were applied to characterize the molecular signals underlying metabolic reprogramming and identify its critical regulators. These pathways were further validated in vivo in rodents and induced human pluripotent stem cell-derived cardiomyocytes.

Results:

We observed that rhythmicity in Tbx18-induced PCs was regulated by aerobic glycolysis. Fibroblasts critically activated metabolic reprogramming and aerobic glycolysis within PCs, and, therefore, regulated pacemaker activity in PCs. The metabolic reprogramming was attributed to the exclusive induction of Aldoc (aldolase c) within PCs after fibroblast-PC integration. Fibroblasts activated the integrin-dependent mitogen-activated protein kinase-E2F1 signal through cell-cell contact and turned on Aldoc expression in PCs. Interruption of fibroblast-PC interaction or Aldoc knockdown nullified electrical activity. Engineered Tbx18-PC tissue sheets were generated to recapitulate the microenvironment within SANs. Aldoc-driven rhythmic machinery could be replicated within tissue sheets. Similar machinery was faithfully validated in de novo PCs of adult mice and rats, and in human PCs derived from induced pluripotent stem cells.

Conclusions:

Fibroblasts drive Aldoc-mediated metabolic reprogramming and rhythmic regulation in SANs. This work details the cellular machinery behind the complex milieu of vertebrate SANs and opens a new direction for future therapy.

Graphical Abstract

Novelty and Significance

What Is Known?

The sinoatrial node (SAN) initiates electric impulses for every heartbeat, and its dysfunction causes a slow heart rate or cardiac arrest.
SAN is characterized by the microenvironment of pacemaker cardiomyocytes (PCs) encased with fibroblasts.
The disintegration of PCs with fibroblasts results in SAN dysfunction.

What New Information Does This Article Contribute?

Fibroblasts induce metabolic reprogramming and activate the PC-specific expression of Aldoc (aldolase c) within SANs through integrin-dependent cell contact.
The activation of aldolase c critically maintains intrinsic aerobic glycolysis in PCs and regulates pacemaker activities.
The engineered tissue sheets are established as an in vitro model of the SAN microenvironment.
From the induced rat PCs and engineered tissue sheet models to human PCs and in vivo animal experiments, we observed fibroblasts induce aerobic glycolysis and regulate rhythmicity by upregulating Aldoc in PCs. In addition to explaining the mechanisms of SAN failure, Aldoc-driven energy replenishment may also be used as a future device-free therapy to restore SAN dysfunction. Establishment of engineered tissue sheets can be used as an in vitro experimental model for the study of SAN diseases and preclinical tests.
In This Issue, see p 1
Meet the First Author, see p 2
Editorial, see p 21
The sinoatrial node (SAN) initiates electric impulses for every heartbeat to maintain life. Its dysfunction causes a slow heart rate, insufficient blood supply, and detrimental consequences such as cardiac arrest.1 In contrast to atrial or ventricular cardiac tissue, the SAN consists of a network of pacemaker cardiomyocytes (PCs) encased with abundant fibroblasts and a heterogeneous connective tissue microenvironment.2–5 This unique structure of SANs is well conserved across vertebrate species.5–7 The microenvironment, through the integration of PCs, mesenchymal lineages (including fibroblasts), and extracellular matrix (ECM) organization, is required for the rhythmic activity of SANs during embryogenesis.4 Failure of ECM organization and likely fibroblast integration results in electrical dysfunction of SANs.4 At the other extreme, extensive fibrosis of the SAN also leads to pacemaker failure.8,9 Alteration of the microenvironment underlies the pathogenesis of SAN disorders. Although the molecular mechanisms underlying the ability of individual PCs to generate rhythmic electrical impulses have been well studied,10,11 the biological process behind the microenvironmental niche in SANs, especially fibroblast-PC interactions, remains poorly understood.
The SAN is tiny with a paucity of PCs.3,12 Operable cell or tissue models are either generally lacking or difficult to handle.10 To study functional interactions within the microenvironment, recapitulation of the complex structure of the PCs and fibroblasts within the SAN is necessary, but such models have yet to be developed.13 This presents a barrier to studying the biological function of this critical tissue and obtaining models of SAN diseases.10 Recently, induced PCs, generated by Tbx18 (T-box transcription factor 18) transduction or biomaterial, recapitulated not only electrical and morphological phenotypes, but also the metabolic properties of native SAN cardiomyocytes.14–16 It is possible that Tbx18-induced PCs (Tbx18-PCs), considered a replacement for native PCs, might be used to establish engineered models to study unknown SAN biological processes, especially intercellular functional interactions within the microenvironment.17
In this study, we used Tbx18-PCs and engineered tissue to explore how fibroblasts regulate the functional integrity of SANs. Fibroblasts drove PC-specific expression of Aldoc (aldolase c, an enzyme involved in glycolysis metabolism) through integrin-dependent cell contact. This machinery critically maintained intrinsic aerobic glycolysis in PCs and regulated pacemaker activities. Aldoc-mediated rhythmic activity was faithfully validated in an in vitro engineered model, in vivo in mice, and in human-induced pluripotent stem cell-derived cardiomyocytes. These findings highlight the importance of the SAN microenvironment in determining its energy metabolism and rhythmicity. Moreover, tissue engineered with Tbx18-PCs could be a feasible in vitro platform to study SAN physiology.

Methods

A detailed, expanded methods section is provided in the Supplemental Material. The Institutional Animal Care and Use Committee at Taipei Veterans General Hospital approved all animal experiments.

Data Availability

All data supporting the findings of this study are available within the article and its Supplemental Material. The whole transcriptome analysis data set (Figure 1A and Figure 3A) is provided in the Sequence Read Archive data at NCBI (PRJNA743181 and PRJNA743409).
Figure 1. Aldoc (aldolase c)-driven glycolysis regulation and pacemaker rhythmicity. A, Heatmap of differential gene expression between Tbx18 (T-box transcription factor 18) pacemaker cardiomyocytes (PCs) and control ventricular cardiomyocytes (VMs). B, Canonical pathways. C, The expression levels of the key metabolic genes in glycolysis between Tbx18-PCs and control-VMs. Upper: The key metabolic genes (red color) and metabolites in the glycolysis process. In the lower part, the enzymes involved in glycolysis mostly increased, and only Aldoc was downregulated. A–C, n=4 for each group. *P<5.0×10−2 (Data 1). D, The expression of metabolic genes in glycolysis determined by real-time PCR (Hk1 [hexokinase-1], control-VMs, n=7; Tbx18-PCs, n=8; Hk2 [hexokinase-2], control-VMs, n=5; Tbx18-PCs, n=7; Aldolase a and Aldoc, control-VMs, n=7; Tbx18-PCs, n=12). E, Seahorse glycolysis stress test. Left: Representative curves from the experiments. Glycolysis was persistently lower in Tbx18-PCs than in control-VMs. The right part shows the measurements including basal glycolysis, % proton efflux rate (PER) and compensatory glycolysis. control-VMs, n=18; Tbx18-PCs, n=14. F, The levels of glycolysis metabolites determined by mass spectrometry. The highest decrease was observed in the relative levels of glyceraldehyde 3-phosphate (G3P) and dihydroxyacetone phosphate (DHAP) to those of control-VMs. G, The reduction ratio of metabolites in Tbx18-PCs with reference to control-VMs. The reduction ratio (%)=100×(Tbx18-PC metabolites−control-VM metabolites/control-VM metabolites). The reduction in DHAP and G3P levels reached a nadir in comparison to the other metabolites. F and G, control-VMs, n=3; Tbx18-PCs, n=4. H, Lactate levels in PCs determined by a colorimetric assay. n=7 for each group. I, Modulation of glycolysis changed pacemaker phenotypes, including potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4 (Hcn4) expression and beating rate. Supplementation of active glycolysis metabolites with sodium pyruvate (1 mmol/L) increased Hcn4 expression (Tbx18-PCs, n=10; Tbx18-PCs+sodium pyruvate, n=7; Tbx18-PCs+2-DG, n=11,) and beating rates (Tbx18-PCs, n=7; Tbx18-PCs+sodium pyruvate, n=8; Tbx18-PCs+2-DG, n=7). 2-DG inhibits glycolysis by competitively inhibiting the production of glucose-6-phosphate from glucose at the phosphoglucoisomerase level. Treatment with 2-DG (5 mmol/L) decreased Hcn4 levels and beating rate. J, Adenoviral vector-mediated overexpression of Aldoc in Tbx18-PCs (Tbx18-PCs-control, n=8; Tbx18-PCs-Aldoc, n=8). K, Aldoc overexpression increased the electrical firing rate (Tbx18-PCs-control, n=17; Tbx18-PCs-Aldoc, n=19) and Hcn4 expression (n=8 for each group). Representative tracing of the electrical firing rate in the Aldoc-overexpression and control vector groups is shown in the left. P value determined by a 2-tailed t test for all except I, determined by 1-way ANOVA.
Figure 2. Fibroblasts drive Aldoc (aldolase c)-mediated glycolysis adaptation in pacemaker cardiomyocytes. A, Glycolysis stress test in Tbx18 (T-box transcription factor 18) pacemaker cardiomyocytes (PCs) and cocultures. Coculture with fibroblasts improved glycolysis, in comparison to the single culture of Tbx18-PCs. The left shows representative curves from the Seahorse glycolysis test for Tbx18-PCs and cocultures (Tbx18-PCs, n=10; coculture, n=12). B, Mitochondrial stress test in Tbx18-PCs and cocultures. The left shows the representative curves from the mitochondrial stress test. Measurements of basal respiration, spare respiratory capacity, proton leak and ATP production did not differ between the 2 groups. Tbx18-PCs, n=11; coculture, n=12. C, The levels in glycolysis metabolites determined by mass spectrometry were compared between Tbx18-PCs and cocultures (relative expression to control-VMs). After cocultures, most of the metabolites increased. However, G3P and dihydroxyacetone phosphate (DHAP) levels exhibited the greatest changes (>2-fold, red arrow). *P<5.0×102 Tbx18-PCs vs cocultures; Tbx18-PCs, n=4; coculture, n=3. D, The level of DHAP determined by ELISA in Tbx18-cocultures was higher than that in Tbx18-PCs (Tbx18-PCs, n=10; coculture, n=11). E, Aldoc transcripts increased in the cocultures with fibroblasts compared with the level in Tbx18-PCs alone (Tbx18-PCs, n=12; coculture, n=13). F, Coculture increased beating rates compared with those of single Tbx18-PC cultures (Tbx18-PCs, n=26; coculture, n=37). G, Coculture also increased the expression of distinct pacemaker genes (Hcn4 [potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4], Cx45 [connexin 45]) compared with Tbx18-PCs alone (Tbx18-PCs, n=9; coculture, n=10). H, The level of the end-product of glycolysis (lactate) represents the active status of glycolysis. The separate-coculture of Tbx18-PCs and Tbx18-transduced fibroblasts was not associated with an increased level of lactate. Only contact-coculture containing both Tbx18-PCs and fibroblasts was associated with increased lactate levels. P value determined by 1-way ANOVA with an least significant difference (LSD) post hoc test. I, Representative western blot images of Aldoc expression. No Aldoc expression was noted in fibroblasts with GAPDH as a protein control (n=4). However, Aldoc expression was observed in neonatal cardiomyocytes (ventricular cardiomyocytes [VMs], n=4). P value determined by Mann-Whitney test (A) or a 2-tailed t test (B–G). OCR indicates oxygen consumption rate.
Figure 3. Fibroblast-pacemaker interaction to regulate intrinsic expression of Aldoc (aldolase c) in pacemaker cardiomyocytes. A, Heatmap of differential gene expression in cardiomyocytes isolated from single Tbx18 (T-box transcription factor 18) pacemaker cardiomyocyte (PC) cultures and fibroblast cocultures (Tbx18-PCs, single culture, n=4; coculture, n=3). B, Transcript levels of the key enzymes involved in glycolysis between cocultures and single cultures of Tbx18-PCs. *P<5.0×10−2 (Supplemental Data S4). The expression of Aldoc was increased. C, The canonical pathways determined by IPA analysis. The detailed genes within these pathways, as shown in Table S3, revealed critical regulation of ECM receptors and their downstream signals after coculture. D, The transcript levels of ECM-binding receptors. Compared with the other integrin subunits, Itgb1 levels were the most abundant and significantly increased after coculture (Tbx18-PCs, single culture, n=4; coculture, n=3). E, Aldoc expression in Tbx18-PC cocultures decreased after treatment with the Itgb1 inhibitory antibody (IgG control, n=10; Itgb1 antibody, n=11). F, Representative Western blot of total and phosphorylated AKT showing no difference between Tbx18-PC single cultures and cocultures. G, Aldoc expression after treatment with a PI3K inhibitor (Wortmannin, n=5 for each group). H, Representative Western blot of total and phosphorylated ERK and p38-MAPK (mitogen-activated protein kinase) showing increased total and phosphorylated p38-MAPK after coculture with fibroblasts. I, Reduced Aldoc expression after treatment with a p38-MAPK inhibitor (control, n=9; SB203580, n=9). J, Representative western blot of total and phosphorylated Rb and E2F1 showing increased total and phosphorylated Rb and E2F1 after coculture with fibroblasts. K, Promoter binding site prediction for Aldoc determined using AnimalTFDB version 3.0 (http://bioinfo.life.hust.edu.cn/AnimalTFDB/). The prediction showed three E2F1 binding sites within the promotor of Aldoc. L, Knockdown of p38-MAPK, Rb, and E2F1 downregulated Aldoc expression (nontarget, n=13; Rb siRNA, n=12; E2F1 siRNA, n=13; p38-MAPK siRNA, n=13). Treatment with p38-MAPK, Rb, and E2F1 siRNAs reduced the expression of the corresponding target genes, as shown in Figure S17. M, Intraperitoneal injection of the Itgb1 inhibitory antibody decreased Aldoc expression in mouse SANs (IgG control, n=6; Itgb1 antibody, n=8). All P values determined by a 2-tailed t test. Statistical analyses of Western blotting results (F, H, and J) and uncut blot data are provided in Figure S11 through S16.

Statistical Analysis

Statistical analysis was performed with GraphPad Prism version 8.3.0 (GraphPad Software, CA). Data are expressed as the mean±SD. The Shapiro-Wilks normality test was used to determine the use of Student t test, 1-way ANOVA, or Mann Whitney test as appropriate. Repeated measures ANOVA with the least significant difference (LSD) post hoc test was used when repeated measures were necessary. Multiple comparison correction was performed using the false discovery rate control approach and the Benjamini–Hochberg method when multiple comparisons of different variants were needed.18 A P value <0.05 was considered statistically significant.

Results

Glycolysis Metabolism Regulated Rhythmicity in Tbx18-PCs

We hypothesized that a cell-specific biological process of PCs, which is different from that of quiescent ventricular cardiomyocytes (VMs), would underlie the tailored integration of fibroblasts and PCs for functional integrity.4,19 Therefore, Tbx18-PCs were selected as the cell model.14,20 The differential biological processes between PCs and VMs were explored. Whole-transcriptome expression was compared between Tbx18-PCs and control-VMs (Figure 1A, Supplemental Data S1).14,20 Glucose metabolism and glycolysis accounted for the top canonical pathways (Figure 1B, Table S1, and Supplemental Data S2) and were predominant in gene ontology analysis (Table S2 and Supplemental Data S3). Glucose is metabolized to pyruvate via a complex enzyme network (Figure 1C). The metabolic genes involved in glycolysis were mostly increased in Tbx18-PCs. Aldoc was an exception, as its transcripts significantly decreased (Figure 1C). The genes in other metabolic pathways, including the tricarboxylic acid cycle cycle, pentose phosphate pathway, pyruvate oxidation, and fatty acid metabolism were either not different or slightly increased between Tbx18-PCs and control-VMs (Figure S1). The differential change in Aldoc transcripts between Tbx18-PCs and control-VMs was confirmed via real-time PCR (Figure 1D).
To correlate functional changes in transcriptome expression, glycolysis and mitochondrial function in Tbx18-PCs were further analyzed using Seahorse functional assays. Glycolysis activity, including basal and compensatory glycolysis, and proton efflux rate in Tbx18-PCs were lower than in control-VMs (Figure 1E). Mitochondrial function, including basal respiration, proton leak, and ATP production, did not differ between the 2 groups (Figure S2). The downregulation of glycolysis was correlated with the reduced levels of Aldoc and suggested that the increased expression of metabolic genes other than Aldoc was likely compensatory. To confirm this idea, we performed metabolomics analysis via liquid chromatography–mass spectrometry to comprehensively delineate the metabolite levels of the aforementioned pathways in Tbx18-PCs and control-VMs.
The levels of tricarboxylic acid cycle cycle metabolites, energy molecules (eg, ATP and NADH), pyruvate conversion metabolites, and pentose phosphate pathway metabolites were mostly not statistically different between Tbx18-PCs and control-VMs (Figure S3). Only within the glycolysis process, almost all metabolites decreased. A maximal decrease was observed in the levels of glyceraldehyde-3-phosphate (G3P) and dihydroxyacetone phosphate (DHAP, Figure 1F), which reached the nadir of all glycolysis metabolites (Figure 1G). Aldoc catalyses reversible aldolase cleavage of fructose 1,6-bisphosphate to DHAP and G3P (Figure 1C).21 The critical reduction in DHAP and G3P levels was consistent with the reduction in Aldoc expression. The reduction in lactate levels (end-product of glycolysis) was further confirmed by a colorimetric assay (Figure 1H). These results reflected low glycolysis activity in the single culture of PCs and demonstrated the key role of reduced Aldoc expression in determining glycolysis status.
Whether glycolysis activity in PCs has functional implications must be determined. Supplementation with sodium pyruvate to replenish the energy supply from glycolysis in Tbx18-PCs increased the beating rates of PCs and potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4 (Hcn4) expression (distinct ion channels that initiate electrical impulses in PCs, Figure 1I). Treatment with 2-deoxy-D-glucose to inhibit glycolysis decreased both beating rates and Hcn4 expression in Tbx18-PCs (Figure 1I). Considered a critical hub in the glycolysis process, adenoviral vector-mediated overexpression of Aldoc was performed to reverse Aldoc expression in Tbx18-PCs (Figure 1J). The increased expression of Aldoc improved beating rates and Hcn4 expression (Figure 1K). These results indicate that reduced Aldoc levels and glycolysis status dysregulated PC rhythmicity.

Fibroblasts Drive Glycolysis Adaptation Through Aldoc in Tbx18-PCs

Next, we determined whether fibroblasts regulate pacemaker rhythm through intrinsic glycolysis within Tbx18-PCs. Coculture with fibroblasts improved global glycolysis function, including basal and compensatory glycolysis, as well as the proton efflux rate, compared with Tbx18-PCs alone (Figure 2A). Mitochondrial function (oxidative phosphorylation), including basal respiration, spare respiratory capacity, proton leak, and ATP production did not differ between the 2 groups (Figure 2B). Based on metabolomics analysis, compared with the drastic decrease in G3P and DHAP in the single culture of Tbx18-PCs, coculture with fibroblasts significantly increased G3P and DHAP, as well as fructose 1,6-bisphosphate, through the reverse reaction of aldolase from DHAP (Figure 2C).22 This led to the upregulation of downstream metabolites, such as 2-phosphoglyceric acid and phosphoenolpyruvate. The levels of tricarboxylic acid cycle cycle metabolites, energy molecules (eg, ATP and NADH), pyruvate conversion metabolites, and pentose phosphate pathway metabolites were mostly marginally increased or not statistically different between Tbx18-PCs and cocultures (Figure S4). The increased levels of DHAP were validated by an ELISA (Figure 2D).
Aldoc expression in the cocultures was higher than that in the single PC cultures (Figure 2E), thus supporting the notion that Aldoc expression underlies the increase in G3P and DHAP levels. In addition, coculture with fibroblasts was associated with better pacemaker phenotypes such as beating rate (Figure 2F), as well as with the expression of PC-specific genes (Hcn4 and Cx45 [connexin 45], Figure 2G). Spontaneous local Ca2+ release events are a hallmark of automaticity in PCs.23,24 Spontaneous local Ca2+ release events could be observed in Tbx18-PC cocultures. The inhibition of Aldoc by the Aldoc siRNAs decreased both spontaneous local Ca2+ release events and oscillating calcium transients (Figure S5). The spontaneous local Ca2+ release event period, as observed in the Tbx18-PC cocultures, was linearly correlated with the cycle length of oscillating calcium transients (Figure S5). Overall, these findings suggest that Aldoc regulates both membrane and calcium clocks within PCs.
The improvement in glycolysis was related to the intrinsic regulation of PCs but not to contamination of fibroblasts. First, lactate levels increased in contact cocultures, which supported the improvement of glycolysis function after coculture with fibroblasts (Figure 2H). However, if we performed a separate coculture (coculture of fibroblasts and PCs, but PCs and fibroblasts were separated by porous membranes), the levels of lactate did not increase. Lactate levels in the single culture of fibroblasts were also low. These findings indicate that the microenvironment contributed to the improvement in glycolytic activity through contact between fibroblasts and PCs. Moreover, we did not observe any Aldoc protein expression in fibroblasts (Figure 2I and Figure S6). These results indicate that the improvement in glycolysis was due to intrinsic regulation of Aldoc within PCs.
The regulatory enzymes of glycolysis in Tbx18-PC cocultures were different from those in cocultures of control-VMs and fibroblasts (Figure S7). In contrast to increased Aldoc expressions within Tbx18-PCs, fibroblasts increased the transcripts of aldolase a in control-VMs after coculture. Therefore, we observed that glycolysis function and DHAP levels improved in both Tbx18-PCs and control-VMs after coculture with fibroblasts (Figure S7).

Fibroblasts Switched on Aldoc Expression in PCs Through Integrin-Dependent Signals

The mechanisms by which fibroblasts regulate Aldoc expression in PCs were further explored. After coculture with fibroblasts, PCs were isolated via cell sorting (Figure S8). The whole transcriptome expression in isolated PCs from PC-fibroblast cocultures was compared with that from PCs in single PC cultures (Figure 3A, Supplemental Data S4). The analysis of glycolysis-related genes revealed that the highest transcriptional changes (4.3-fold increment) were observed at Aldoc levels compared with the other glycolysis enzymes (Figure 3B). Other metabolic genes related to pyruvate oxidation, tricarboxylic acid cycle, the pentose phosphate pathway, and fatty acid metabolism were either minimally or not statistically different (Figure S9). Again, Aldoc was the key enzyme critically regulated by fibroblasts. The relevant pathways related to metabolic/energetic regulation of automaticity, mainly those within the calcium clock, were analyzed (Figure S10).24,25 The spontaneous electrical activity of PCs, especially calcium clock, is driven by a cAMP-mediated phosphorylation, which critically relies on the balance between the cAMP production by adenylyl cyclases and degradation by cyclic nucleotide Pde (phosphodiesterases).24,25 The coculture with fibroblasts decreased the expression of Pde4a (phosphodiesterase) and might regulate cAMP-mediated protein phosphorylation and the calcium clock in PCs.26
We performed Ingenuity Pathway Analysis to explore the pathways to potentially regulate Aldoc expression, including canonical pathways and gene ontology (Figure 3C, Tables S3 and S4, Supplemental Data S5 and S6). Within canonical pathways (Table S3), those related to ECM (eg, collagen and laminin), relevant surface receptors (integrins) and their downstream signals (PI3K [phosphoinositide 3-kinases]/Akt and MAPK [mitogen-activated protein kinase]) were repeatedly observed. These results were compatible with the finding that direct contact between fibroblast-PCs and the ECM is critical to drive metabolic reprogramming in PCs. Therefore, we comprehensively analyzed integrins, which are the predominant ECM-binding receptors in cardiomyocytes (Figure 3D).27 The Itgb1 (integrin subunit β1) transcripts were the most abundant and increased significantly. We further treated Tbx18-PC coculture with an Itgb1 inhibitory antibody, which decreased integrin activation, Aldoc expression (Figure 3E), and the beating rate (control versus Itgb-1 antibody: 140.4±92.0 bpm, n=23 versus 20.6±55.7 bpm, n=16, P=1.2×10−5). This indicates that fibroblasts activate Aldoc and glycolysis activity through integrin-dependent signals.
The common downstream signals of integrins within cardiomyocytes include PI3K/Akt and MAPK (p38 and ERK).27,28 The expression of phosphorylated and total Akt did not change between cocultures and single cultures of PCs (Figure 3F and Figure S11A and S12). Treatment with a PI3K inhibitor (wortmannin) in PC cocultures increased Aldoc expression (Figure 3G). These results indicate that PI3K/Akt activity did not activate Aldoc expressions after coculture. Considering MAPK pathways, ERK activation was suppressed after coculture as p-ERK (phosphorylated ERK) was lower in cocultures than in the single culture of PCs (Figure 3H and Figure S11B and S13). Instead, phosphorylated and total p38-MAPK increased after coculture (Figure 3H and Figure S11C and S14). Treatment of cocultures with a p38-MAPK inhibitor (SB203580) decreased Aldoc expression, suggesting that p38-MAPK activation is a downstream signal of integrins (Figure 3I) to increase Aldoc expression. The p38-MAPK activation induces the dissociation of Rb and E2F1, and increases E2F1-driven transcriptional activity.29 Increased expression of Rb, phosphorylated Rb, and E2F1 was observed in cocultures (Figure 3J, Figure S11D and S11E, S15, and S16). There are multiple E2F1 binding sites on the promotor region of Aldoc, suggesting that E2F1 is a transcriptional regulator of Aldoc (Figure 3K). Inhibition of p38-MAPK, Rb, and E2F1 via siRNA decreased Aldoc expression (Figure 3L and Figure S17). We further performed an in vivo experiment to clarify the link between Aldoc expression and integrin-dependent signaling. Mice that received an intraperitoneal injection of the Itgb1 inhibitory antibody (a functional blocking antibody) had lower Aldoc expression in SANs than control mice (Figure 3M). Overall, these results suggest that fibroblasts induce Aldoc expression in PCs through β1-integrin activation and downstream p38-MAPK/E2F1 signaling.

Engineered Tbx18-PC Tissue Sheets Recapitulated Aldoc-Driven Rhythmic Machinery

The microenvironment with fibroblasts is essential for the functional integrity of pacemaker tissue. We further constructed an in vitro Tbx18-PC tissue sheet to mimic the 3-dimensional microenvironment of in vivo SANs to study the regulatory role of Aldoc in pacemaker rhythmicity. The PC tissue sheet was induced by re-expression of Tbx18 in an engineered tissue, which was constructed by mixed culture of VMs and fibroblasts with Matrigel (Figure 4A). Compared with control tissue sheets (GFP expression), re-expression of Tbx18 induced spontaneous electrical firing that was recorded with a microelectrode array (Figure 4B). A sympathomimetic drug (epinephrine, α- and β-receptor sympathetic agonist) increased the beating rate of Tbx18-PC tissue sheets (Figure 4C). Through immunofluorescence staining, PCs with distinct pacemaker ion channels (HCN4, Figure 4D and Figure S18) and Cx45 (Figure S19) were observed in Tbx18-PC tissue sheets but not in controls. PC tissue sheets also had higher expression of PC-specific genes (Hcn4 and Cx45) than control tissue sheets (Figure 4E). These findings suggest that engineered Tbx18-PC tissue sheets recapitulated the phenotypes of a native SAN. Aldoc-driven rhythmic machinery within a 3-dimensional microenvironment was further explored. Aldoc expression in PC tissue sheets was higher than controls (Figure 4F). Treatment of PC tissue sheets with Aldoc siRNAs reduced Aldoc expression (Figure S20) and spontaneous electrical activity (Figure 4G). Engineered Tbx18-PC tissue sheets recapitulated the microenvironment of de novo SAN tissues and suggested that the Aldoc-driven glycolysis machinery regulates PC rhythmicity in a 3-dimensional microenvironment.
Figure 4. Engineered Tbx18 (T-box transcription factor 18)-pacemaker tissue sheets recapitulate Aldoc (aldolase c)-driven rhythmic machinery. A, Immunofluorescence staining of a Tbx18-pacemaker cardiomyocyte (PC) tissue sheet. Cardiomyocytes are interspersed with fibroblasts (n=4, thickness of 28.3±10.9 μm). The surface of the engineered Tbx18-PC tissue sheets was mostly covered by fibroblasts. DAPI (nucleus): blue; vimentin (a filament protein in fibroblasts): green; ACTN2 (α-sarcomeric actinin, cardiomyocyte marker): red. B, Electrical recordings of spontaneous firing in Tbx18-PC and control tissue sheets determined by microelectrode array (MEA; control, n=8; Tbx18-PC tissue sheet, n=7). C, The autonomic response of Tbx18-PC tissue sheets. Treatment with the sympathomimetic drug (epinephrine, 0.15 µg/mL, agonist of alpha and beta receptors) increased beating rates in Tbx18-PC tissue sheets compared with rates in controls. There were minimal changes in the beating rate in the control tissue sheets (control, n=6; Tbx18-PC tissue sheet, n=6). D, Fluorescence staining of HCN4 (potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4) in Tbx18-PC tissue sheets. No HCN4 expression was observed in controls (Figure S18). DAPI: blue; HCN4: green; ACTN2: red. E, Hcn4 and Cx45 (connexin 45) transcripts increased in Tbx18-PC tissue sheets compared with levels in controls. Hcn4, control, n=8; Tbx18-PC tissue sheets, n=5; Cx45, control, n=8; Tbx18-PC tissue sheets, n=7. F, Aldoc expression was higher in Tbx18-PC tissue sheets than in controls (n=4 for both groups). G, Knockdown of Aldoc by siRNAs in Tbx18-PC tissue sheets downregulated Aldoc transcripts (Figure S20) and decreased the electrical firing rate of PC tissue sheets (nontarget siRNA, n=11; Aldoc siRNA, n=10). The left shows a representative MEA tracing of nontarget siRNA and Aldoc siRNAs on Tbx18-PC tissue sheets. P value determined by a 2-tailed t test (B–G).

The Regulation of In Vivo Pacemaker Rhythms by Aldoc in Vertebrates

The regional distribution of Aldoc and its physiological function in the hearts of vertebrates were further explored. In rats and mice, through immunofluorescence staining, Aldoc expression was observed exclusively within SANs but not in the atrium or ventricle (Figure 5A). This finding was consistent with the abundance of Aldoc transcripts in rat SANs, while Aldoc expression was almost undetectable in the atria and ventricles (Figure 5B).30 We further performed in vivo knockdown of Aldoc in mouse SANs to determine whether Aldoc regulates the pacemaker activity of SANs. The efficiency of AAV9 (adeno-associated virus 9) Aldoc siRNAs in reducing Aldoc expression was first confirmed via in vitro transduction of mouse cardiomyocytes (Figure S21). Then, AAV9 Aldoc siRNAs were delivered into the pericardial recess around mouse SANs through a mini-thoracotomy and reached ≈80% transduction efficiency (Figure S22). The in vivo Aldoc expression in the mouse SAN was successfully reduced (Figure 5C and 5D). Mice that received Aldoc siRNAs had a lower spontaneous heart rate than those that received the scrambled controls (Figure 5E and 5F). In addition, the responses to epinephrine were nullified in mice that received Aldoc siRNAs (Figure 5F). Accordingly, the expression of Aldoc within the SAN might not only drive glycolysis machinery but also regulate in vivo PC rhythmicity.
Figure 5. Regulation of in vivo pacemaker rhythms by Aldoc (aldolase c) in vertebrates. A, Aldoc expression in adult rat sinoatrial nodes (SANs) determined by immunofluorescence staining. Aldoc expression was observed in adult rat SANs (animal=3) but not in adult rat ventricles (animal=2). Aldoc: red; HCN4 [potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4]: green; DAPI: blue. B, Aldoc transcripts determined by real-time PCR, suggesting dominant expression of Aldoc in the SAN but not in atrial or ventricular tissues. SAN, n=9; atrium, n=4; ventricle, n=8. P value determined by one-way ANOVA with an least significant difference (LSD) post hoc test. C and D, In vivo Aldoc expression in the mouse SAN after transduction with AAV9-Aldoc siRNAs. Regional Aldoc expression within the mouse SAN was best evaluated by immunofluorescence staining, as the SAN could be precisely localized by the presence of HCN4 channels. A representative image is shown in C. Aldoc: gray; HCN4: red; DAPI: blue. D, The fluorescence intensity of Aldoc was significantly reduced after transduction with AAV9 siRNA (AU: arbitrary unit). P value by a 2-tailed t test. Three animals were used for both groups. E, Representative ECG tracings of mice receiving AAV9-Aldoc or scramble siRNAs. In vivo Aldoc knockdown within SANs led to a slower heart rate compared with that in control mice. The intraperitoneal injection of epinephrine (2.5 μg, Epi) increased the heart rate in control mice but not in Aldoc knockdown mice. The results are provided in F. Scramble, n=8; Aldoc knockdown, n=10. P value by a repeated-measures ANOVA followed by an LSD post hoc test.

Aldoc Regulated Pacemaker Activity in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes

Induced pluripotent stem cell-derived cardiomyocytes (IPS-CMs) were used as a human cell model to study whether Aldoc regulates pacemaker activity in human PCs. HCN4 (+) PCs accounted for ≈10% of IPS-CMs31,32 and were responsible for the rhythmic beating of IPS-CMs.33 Aldoc expression was predominantly observed in human HCN4 (+) PCs (Figure 6A and 6B). Overexpression of Aldoc in IPS-CMs via adenoviral vectors significantly increased Aldoc levels in IPS-CMs (Figure 6C) and the electrical firing rate of IPS-CMs (Figure 6D and 6E). Human PCs share similar machinery in which Aldoc drives metabolic adaptation to regulate pacemaker rhythm, highlighting their translational potential for the study of human SAN physiology or disease prevention.
Figure 6. Aldoc (aldolase c) regulates pacemaker activity in human induced pluripotent stem cell-derived cardiomyocytes. A, Immunofluorescence staining showing Aldoc expression in HCN4 (potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4; +) pacemaker cardiomyocytes among human induced pluripotent stem cell-derived cardiomyocytes (IPS-CMs). Aldoc: gray; HCN4: green; ACTN2: red; DAPI: blue. B, Aldoc levels, calculated by fluorescence intensity, were higher in HCN4 (+) PCs than in HCN4 (−) cardiomyocytes (IPS-CMs, n=35; IPS-PCs, n=23). C, Aldoc levels after transduction with adenoviral Aldoc vectors (control, n=4; Aldoc, n=6). D, Aldoc overexpression in IPS-CMs after treatment with the adenoviral vector was associated with higher electrical firing rates in the microelectrode array (MEA) compared with rates in IPS-CMs transduced with control vectors, as shown in the representative MEA tracing. E, Either at baseline (without epinephrine) or after epinephrine treatment (150 ng/mL), increased Aldoc expression in IPS-CMs drove a higher electrical firing rate than observed in controls; n=7 for both groups. P value determined by a 2-tailed t test (B and C), or a repeated-measures ANOVA followed by an least significant difference (LSD) post hoc test (E).

Discussion

Years after observing the clinical linkage of the microenvironmental niche in SANs with pacemaker failure, the exploration of its biological process remains a rocky terrain.11,34 From the induced rat PCs and engineered tissue sheet models to human PCs and in vivo animal experiments, we found that fibroblasts induce metabolic reprogramming by upregulating Aldoc in PCs through integrin-dependent MAPK-E2F1 signals. This enhanced aerobic glycolysis and regulated the rhythmicity of SANs. Interruption of the fibroblast-PC Aldoc interaction stagnated SAN electrical activity. Aldoc-driven energy replenishment may restore SAN dysfunction and might be a future device-free therapeutic target. Establishment of engineered tissue sheets can hopefully be used as an in vitro experimental model for the study of SAN diseases and preclinical tests.
Fibroblast and PC interactions in SANs are rarely addressed. Only a mathematical model suggests that fibroblasts, as stretch sensors during atrial diastole, raise the spontaneous depolarization rate of PCs.35 Our findings uncovered functional implications of the cellular architecture of SANs showing that fibroblasts drive PC-specific Aldoc expression to modulate cardiac rhythm. Constitutive Aldoc expression has mainly been observed in cancer cells or neuronal cells in the brain and causes cancer progression and neurodegeneration.21,36,37 The physiological roles of Aldoc in the heart are far from clear. The constitutive expression of Aldoc in the atria and ventricles is seen in amphibians but not in mammals.36,38 Aldoc seems inducible only in quiescent cardiomyocytes during hypoxia, which activates glycolysis (anaerobic glycolysis) to compensate for energy deficiency from impaired mitochondrial function.39–41
In SANs, the presence of fibroblast-PC interactions activated integrin-dependent pathways and maintained constitutive Aldoc expression within PCs. Even while the mitochondrial oxidative phosphorylation function remained activated and unchanged (as shown in the Seahorse, whole transcriptome, and metabolomics analyses), Aldoc in PCs activated glycolysis (aerobic glycolysis, Warburg effect)42 and regulated beating rates in SANs. This phenomenon was observed either at rest or after treatment with sympathomimetic drugs to mimic exercise. Aerobic glycolysis is an important energy machinery for proliferative cells, including cancer cells, stem cells, and endothelial cells in the heart.42 The present results extend the physiological implications of aerobic glycolysis from cell proliferation to relentless electrical firing in PCs. Most intriguingly, this machinery needs to be maintained by the microenvironment between fibroblasts and PCs.
The upregulation of Itga5 and Itgb1 after the coculture with fibroblasts in the present work suggests that the integrin heterodimer (α5β1) is a critical receptor that drives fibroblast-induced glycolysis signals. The α5β1 is predominantly a fibronectin receptor, which corresponds to the abundant expression of fibronectin in the sinus node ECM.30 Fibroblast-driven Aldoc-mediated PC regulation is similarly observed across different vertebral PC cells, including rats, mice, and human cells, suggesting its physiological importance in SAN function. The critical role of α5β1 in the fibroblast-SAN interaction, ECM, and microenvironment is worth further investigation. The paucity of connexin expression at fibroblast-PC contacts, including Cx45 and Cx43 (data not shown), might be associated with low electrical conductivity across heterocellular junctions. It remains unclear whether the low conductivity between fibroblasts and PCs might be related to the initiation or organization of rhythmic electrical activity.
Aldoc expression are confined to the central nervous system and is not found in the peripheral nervous system (which innervates the heart or SAN). Aldoc is only detected in Purkinje cells, a specific type of cerebellar neuronal cell within the cerebellum.43,44 In other areas of the central nervous system, Aldoc is not detected within neuronal cells and is only observed in nonneuronal cells (astrocytes, some cells of the pia mater) within the hippocampus and thalamus.43,44 Additionally, in our study, Aldoc was found to be expressed exclusively within the cytoplasm of cardiomyocytes in SANs by immunofluorescence staining. These findings suggest that Aldoc is activated within PCs but not via SAN innervation by nerve terminals.
We could never successfully explore the machinery behind PC and fibroblast interactions without the engineered Tbx18-PC and tissue sheet models. Tbx18-PCs and tissue sheets recapitulate the phenotypes of de novo SANs including distinct genes related to pacemaker functions (Hcn4, Cx45), spontaneous electrical firing, the calcium clock, and responses to autonomic stimulation.14 In addition, metabolic machinery including Aldoc-driven glycolysis and PC rhythmicity was similarly replicated in engineered Tbx18-PCs, native SANs in animals, and PCs among IPS-CMs. This similarity underlies the successful translation of fibroblast-driven glycolysis pathways in induced Tbx18-PCs and their rhythmicity to de novo PCs. The engineered Tbx18-PCs and tissue sheets may represent an in vitro platform to study SAN diseases, at least those related to energy metabolism. The SAN is a complex 3-dimensional structure comprising different structures, such as pacemaker cells, transitional cells, fibroblasts, and autonomic nerve terminals. The present engineered model only partially recapitulated SAN phenotypes. Additionally, it should be noted that not all VMs were transduced with Tbx18 and successfully reprogrammed. The Tbx18 transduction rate was 87.3±10.9% (n=3). The reprogramming rate (ie, the ratio of HCN4 [+] cells within Tbx18-transduced cardiomyocytes) was 25.8±14.7%.
Clinical reports addressing Aldoc in SAN pathologies are not well established. This is probably not a limitation but rather offers new hope to understand the mechanisms of SAN diseases and develop future therapy. For example, age-dependent degeneration is a common cause of SAN dysfunction,11 and a reduction in blood Aldoc expression is observed with aging.45 Exploring the potential role of reduced Aldoc expression in the aging SAN might give birth to a target strategy involving Aldoc replenishment and restoration of SAN failure. This could not only be considered a future device-free therapy but also part of preventive medicine. Regulation of glucose-related cardiac metabolism by diet, lifestyle, exercise, or medication for the primary prevention of cardiac disease is not new.46

Article Information

Author Contributions

P.-C. Chou, C.-H. Weng, and J.-D. Liu performed immunostaining, PCR, in vivo and in vitro cell experiments, and animal experiments; K.-C. Yang performed whole transcriptome analysis. M.-L. Cheng performed metabolomics analysis; R.-B. Yang and Y.-C. Lin performed the integrin and mechanism experiments. B.-C. Shyu assisted in microelectrode array experiments. M. Hsiao and S.-K. Shyue assisted in the gene interference experiments. S.-P. Chen reviewed the results. P.-C. Chou, C.-H. Weng, and C.-M. Liu wrote the article. Y.-F. Hu designed the project and revised the article.

Supplemental Materials

Expanded Methods and Materials
Data Set 1–6
References 14,20,31,33,47–55

Acknowledgments

We thank Wei-Chi Wang for bioinformatics analysis and I-Chien Wu for statistical analysis consultation.

Footnote

Nonstandard Abbreviations and Acronyms

Aldoc
aldolase c
Cx45
connexin 45
DHAP
dihydroxyacetone phosphate
ECM
extracellular matrix
G3P
glyceraldehyde 3-phosphate
Hcn4
potassium/sodium hyperpolarization-activated cyclic nucleotide-gated channel 4
IPS-CMs
induced pluripotent stem cell-derived cardiomyocytes
Itgb1
integrin subunit β1
MAPK
mitogen-activated protein kinase
PCs
pacemaker cardiomyocytes
Pde
phosphodiesterase
Pde4a
phosphodiesterase
p-ERK
phosphorylated ERK
PI3K
phosphoinositide 3-kinase
SAN
sinoatrial node
Tbx18
T-box transcription factor 18
Tbx18-PC
Tbx18-induced PC
VMs
ventricular cardiomyocytes

Supplemental Material

File (circres_circres-2021-320301_supp1.xlsx)
File (circres_circres-2021-320301_supp2.pdf)
File (circres_circres-2021-320301_supp3.xlsx)
File (circres_circres-2021-320301_supp4.xlsx)
File (circres_circres-2021-320301_supp5.pdf)
File (circres_circres-2021-320301_supp6.xlsx)
File (circres_circres-2021-320301_supp7.pdf)
File (circres_circres-2021-320301_supp8.pdf)

References

1.
Epstein AE, DiMarco JP, Ellenbogen KA, Estes NA, Freedman RA, Gettes LS, Gillinov AM, Gregoratos G, Hammill SC, Hayes DL, et al; American College of Cardiology Foundation; American Heart Association Task Force on Practice Guidelines; Heart Rhythm Society. 2012 ACCF/AHA/HRS focused update incorporated into the ACCF/AHA/HRS 2008 guidelines for device-based therapy of cardiac rhythm abnormalities: a report of the american college of cardiology foundation/american heart association task force on practice guidelines and the heart rhythm society. Circulation. 2013;127:e283–e352. doi: 10.1161/CIR.0b013e318276ce9b
2.
Camelliti P, Green CR, LeGrice I, Kohl P. Fibroblast network in rabbit sinoatrial node: structural and functional identification of homogeneous and heterogeneous cell coupling. Circ Res. 2004;94:828–835. doi: 10.1161/01.RES.0000122382.19400.14
3.
Perde FV, Atkinson A, Yanni J, Dermengiu D, Dobrzynski H. Morphological characteristics of the sinus node on postmortem tissue. Folia Morphol (Warsz). 2016;75:216–223. doi: 10.5603/FM.a2015.0087
4.
Bressan M, Henley T, Louie JD, Liu G, Christodoulou D, Bai X, Taylor J, Seidman CE, Seidman JG, Mikawa T. Dynamic cellular integration drives functional assembly of the heart’s pacemaker complex. Cell Rep. 2018;23:2283–2291. doi: 10.1016/j.celrep.2018.04.075
5.
Bleeker WK, Mackaay AJ, Masson-Pévet M, Bouman LN, Becker AE. Functional and morphological organization of the rabbit sinus node. Circ Res. 1980;46:11–22. doi: 10.1161/01.res.46.1.11
6.
Opthof T. The mammalian sinoatrial node. Cardiovasc Drugs Ther. 1988;1:573–597. doi: 10.1007/BF02125744
7.
Kalyanasundaram A, Li N, Hansen BJ, Zhao J, Fedorov VV. Canine and human sinoatrial node: differences and similarities in the structure, function, molecular profiles, and arrhythmia. J Vet Cardiol. 2019;22:2–19. doi: 10.1016/j.jvc.2018.10.004
8.
Monfredi O, Boyett MR. Sick sinus syndrome and atrial fibrillation in older persons - A view from the sinoatrial nodal myocyte. J Mol Cell Cardiol. 2015;83:88–100. doi: 10.1016/j.yjmcc.2015.02.003
9.
Ferrer MI. The etiology and natural history of sinus node disorders. Arch Intern Med. 1982;142:371–372.
10.
Cingolani E, Goldhaber JI, Marbán E. Next-generation pacemakers: from small devices to biological pacemakers. Nat Rev Cardiol. 2018;15:139–150. doi: 10.1038/nrcardio.2017.165
11.
Dobrzynski H, Boyett MR, Anderson RH. New insights into pacemaker activity: promoting understanding of sick sinus syndrome. Circulation. 2007;115:1921–1932. doi: 10.1161/CIRCULATIONAHA.106.616011
12.
Opthof T, de Jonge B, Mackaay AJ, Bleeker WK, Masson-Pevet M, Jongsma HJ, Bouman LN. Functional and morphological organization of the guinea-pig sinoatrial node compared with the rabbit sinoatrial node. J Mol Cell Cardiol. 1985;17:549–564. doi: 10.1016/s0022-2828(85)80024-9
13.
Weinberger F, Mannhardt I, Eschenhagen T. Engineering cardiac muscle tissue: a maturating field of research. Circ Res. 2017;120:1487–1500. doi: 10.1161/CIRCRESAHA.117.310738
14.
Kapoor N, Liang W, Marbán E, Cho HC. Direct conversion of quiescent cardiomyocytes to pacemaker cells by expression of Tbx18. Nat Biotechnol. 2013;31:54–62. doi: 10.1038/nbt.2465
15.
Gu JM, Grijalva SI, Fernandez N, Kim E, Foster DB, Cho HC. Induced cardiac pacemaker cells survive metabolic stress owing to their low metabolic demand. Exp Mol Med. 2019;51:1–12. doi: 10.1038/s12276-019-0303-6
16.
Hu YF, Lee AS, Chang SL, Lin SF, Weng CH, Lo HY, Chou PC, Tsai YN, Sung YL, Chen CC, et al. Biomaterial-induced conversion of quiescent cardiomyocytes into pacemaker cells in rats. Nat Biomed Eng. 2022;6:421–434. doi: 10.1038/s41551-021-00812-y
17.
Grijalva SI, Gu JM, Li J, Fernandez N, Fan J, Sung JH, Lee SY, Herndon C, Buckley EM, Park SJ, et al. Engineered cardiac pacemaker nodes created by TBX18 gene transfer overcome source-sink mismatch. Adv Sci (Weinh). 2019;6:1901099. doi: 10.1002/advs.201901099
18.
Glickman ME, Rao SR, Schultz MR. False discovery rate control is a recommended alternative to Bonferroni-type adjustments in health studies. J Clin Epidemiol. 2014;67:850–857. doi: 10.1016/j.jclinepi.2014.03.012
19.
Furtado MB, Nim HT, Boyd SE, Rosenthal NA. View from the heart: cardiac fibroblasts in development, scarring and regeneration. Development. 2016;143:387–397. doi: 10.1242/dev.120576
20.
Hu YF, Dawkins JF, Cho HC, Marbán E, Cingolani E. Biological pacemaker created by minimally invasive somatic reprogramming in pigs with complete heart block. Sci Transl Med. 2014;6:245ra94. doi: 10.1126/scitranslmed.3008681
21.
Arakaki TL, Pezza JA, Cronin MA, Hopkins CE, Zimmer DB, Tolan DR, Allen KN. Structure of human brain fructose 1,6-(bis)phosphate aldolase: linking isozyme structure with function. Protein Sci. 2004;13:3077–3084. doi: 10.1110/ps.04915904
22.
Choi KH, Shi J, Hopkins CE, Tolan DR, Allen KN. Snapshots of catalysis: the structure of fructose-1,6-(bis)phosphate aldolase covalently bound to the substrate dihydroxyacetone phosphate. Biochemistry. 2001;40:13868–13875. doi: 10.1021/bi0114877
23.
Hu YF, Lee AS, Chang SL, Lin SF, Weng CH, Lo HY, Chou PC, Tsai YN, Sung YL, Chen CC, et al. Biomaterial-induced conversion of quiescent cardiomyocytes into pacemaker cells in rats. Nat Biomed Eng. 2022;6:421–434. doi: 10.1038/s41551-021-00812-y
24.
Lakatta EG, Maltsev VA, Vinogradova TM. A coupled system of intracellular Ca2+ clocks and surface membrane voltage clocks controls the timekeeping mechanism of the heart’s pacemaker. Circ Res. 2010;106:659–673. doi: 10.1161/CIRCRESAHA.109.206078
25.
Yaniv Y, Juhaszova M, Lyashkov AE, Spurgeon HA, Sollott SJ, Lakatta EG. Ca2+-regulated-cAMP/PKA signaling in cardiac pacemaker cells links ATP supply to demand. J Mol Cell Cardiol. 2011;51:740–748. doi: 10.1016/j.yjmcc.2011.07.018
26.
Vinogradova TM, Sirenko S, Lukyanenko YO, Yang D, Tarasov KV, Lyashkov AE, Varghese NJ, Li Y, Chakir K, Ziman B, Lakatta EG. Basal spontaneous firing of rabbit sinoatrial node cells is regulated by dual activation of PDEs (phosphodiesterases) 3 and 4. Circ Arrhythm Electrophysiol. 2018;11:e005896. doi: 10.1161/CIRCEP.117.005896
27.
Israeli-Rosenberg S, Manso AM, Okada H, Ross RS. Integrins and integrin-associated proteins in the cardiac myocyte. Circ Res. 2014;114:572–586. doi: 10.1161/CIRCRESAHA.114.301275
28.
Roux PP, Blenis J. ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev. 2004;68:320–344. doi: 10.1128/MMBR.68.2.320-344.2004
29.
Wang S, Nath N, Minden A, Chellappan S. Regulation of Rb and E2F by signal transduction cascades: divergent effects of JNK1 and p38 kinases. EMBO J. 1999;18:1559–1570. doi: 10.1093/emboj/18.6.1559
30.
Linscheid N, Logantha SJRJ, Poulsen PC, Zhang S, Schrölkamp M, Egerod KL, Thompson JJ, Kitmitto A, Galli G, Humphries MJ, et al. Quantitative proteomics and single-nucleus transcriptomics of the sinus node elucidates the foundation of cardiac pacemaking. Nat Commun. 2019;10:2889. doi: 10.1038/s41467-019-10709-9
31.
Tsai MH, Chiu YT, Chan DZ, Wen CH, Syu SH, Lu HE, Huang CF, Lin YJ, Chang SL, Lo LW, et al. Generation of IBMS-iPSC-021, -022, -023 human induced pluripotent stem cells (IBMSi016-A, IBMSi017-A, and IBMSi018-A) derived from patients with the ALDH2 rs671 polymorphism. Stem Cell Res. 2021;54:102416. doi: 10.1016/j.scr.2021.102416
32.
Chang CW, Kao HKJ, Yechikov S, Lieu DK, Chan JW. An intrinsic, label-free signal for identifying stem cell-derived cardiomyocyte subtype. Stem Cells. 2020;38:390–394. doi: 10.1002/stem.3127
33.
Chiu YT, Tsai MH, Chan DZ, Ko HW, Lu HE, Huang CF, Lin YJ, Chang SL, Lo LW, Huang CY, et al. Generation of IBMS-iPSC-015, -016, -017 human induced pluripotent stem cells (IBMSi013-A, IBMSi014-A, and IBMSi015-A) derived from patients with atrial fibrillation. Stem Cell Res. 2021;54:102419. doi: 10.1016/j.scr.2021.102419
34.
Bañuls MP, Alvarez A, Ferrero I, Zapata A, Ardavin C. Cell-surface marker analysis of rat thymic dendritic cells. Immunology. 1993;79:298–304.
35.
Kohl P, Kamkin AG, Kiseleva IS, Noble D. Mechanosensitive fibroblasts in the sino-atrial node region of rat heart: interaction with cardiomyocytes and possible role. Exp Physiol. 1994;79:943–956. doi: 10.1113/expphysiol.1994.sp003819
36.
Shiokawa K, Kajita E, Hara H, Yatsuki H, Hori K. A developmental biological study of aldolase gene expression in xenopus laevis. Cell Res. 2002;12:85–96. doi: 10.1038/sj.cr.7290114
37.
Yu Q, Huang Q, Du X, Xu S, Li M, Ma S. Early activation of Egr-1 promotes neuroinflammation and dopaminergic neurodegeneration in an experimental model of Parkinson’s disease. Exp Neurol. 2018;302:145–154. doi: 10.1016/j.expneurol.2018.01.009
38.
Langellotti S, Romano M, Guarnaccia C, Granata V, Orrù S, Zagari A, Baralle FE, Salvatore F. A novel anti-aldolase C antibody specifically interacts with residues 85-102 of the protein. MAbs. 2014;6:708–717. doi: 10.4161/mabs.28191
39.
Yang H, Shao N, Holmstrom A, Zhao X, Chour T, Chen H, Itzhaki I, Wu H, Ameen M, Cunningham NJ, et al. Transcriptome analysis of non-human primate induced pluripotent stem cell-derived cardiomyocytes in 2D monolayer culture versus 3D engineered heart tissue. Cardiovasc Res. 2020;117:2125–2136. doi: 10.1093/cvr/cvaa281
40.
Jian B, Wang D, Chen D, Voss J, Chaudry I, Raju R. Hypoxia-induced alteration of mitochondrial genes in cardiomyocytes: role of Bnip3 and Pdk1. Shock. 2010;34:169–175. doi: 10.1097/SHK.0b013e3181cffe7d
41.
Bertero E, Maack C. Metabolic remodelling in heart failure. Nat Rev Cardiol. 2018;15:457–470. doi: 10.1038/s41569-018-0044-6
42.
Chen Z, Liu M, Li L, Chen L. Involvement of the warburg effect in non-tumor diseases processes. J Cell Physiol. 2018;233:2839–2849. doi: 10.1002/jcp.25998
43.
Thompson RJ, Kynoch PA, Willson VJ. Cellular localization of aldolase C subunits in human brain. Brain Res. 1982;232:489–493. doi: 10.1016/0006-8993(82)90294-3
44.
Walther EU, Dichgans M, Maricich SM, Romito RR, Yang F, Dziennis S, Zackson S, Hawkes R, Herrup K. Genomic sequences of aldolase C (Zebrin II) direct lacZ expression exclusively in non-neuronal cells of transgenic mice. Proc Natl Acad Sci U S A. 1998;95:2615–2620. doi: 10.1073/pnas.95.5.2615
45.
Spinetti G, Sangalli E, Specchia C, Villa F, Spinelli C, Pipolo R, Carrizzo A, Greco S, Voellenkle C, Vecchione C, et al. The expression of the BPIFB4 and CXCR4 associates with sustained health in long-living individuals from Cilento-Italy. Aging (Albany NY). 2017;9:370–380. doi: 10.18632/aging.101159
46.
Krist AH, Davidson KW, Mangione CM, Barry MJ, Cabana M, Caughey AB, Donahue K, Doubeni CA, Epling JW, Kubik M, et al. Behavioral counseling interventions to promote a healthy diet and physical activity for cardiovascular disease prevention in adults with cardiovascular risk factors: us preventive services task force recommendation statement. JAMA. 2020;324:2069–2075. doi: 10.1001/jama.2020.21749
47.
Kizana E, Chang CY, Cingolani E, Ramirez-Correa GA, Sekar RB, Abraham MR, Ginn SL, Tung L, Alexander IE, Marbán E. Gene transfer of connexin43 mutants attenuates coupling in cardiomyocytes: novel basis for modulation of cardiac conduction by gene therapy. Circ Res. 2007;100:1597–1604. doi: 10.1161/CIRCRESAHA.106.144956
48.
Sekar RB, Kizana E, Cho HC, Molitoris JM, Hesketh GG, Eaton BP, Marbán E, Tung L. IK1 heterogeneity affects genesis and stability of spiral waves in cardiac myocyte monolayers. Circ Res. 2009;104:355–364. doi: 10.1161/CIRCRESAHA.108.178335
49.
Neuss M, Regitz-Zagrosek V, Hildebrandt A, Fleck E. Isolation and characterisation of human cardiac fibroblasts from explanted adult hearts. Cell Tissue Res. 1996;286:145–153. doi: 10.1007/s004410050683
50.
Tsai YC, Tsai TH, Chang CP, Chen SF, Lee YM, Shyue SK. Linear correlation between average fluorescence intensity of green fluorescent protein and the multiplicity of infection of recombinant adenovirus. J Biomed Sci. 2015;22:31. doi: 10.1186/s12929-015-0137-z
51.
Yang KC, Ku YC, Lovett M, Nerbonne JM. Combined deep microRNA and mRNA sequencing identifies protective transcriptomal signature of enhanced PI3Kα signaling in cardiac hypertrophy. J Mol Cell Cardiol. 2012;53:101–112. doi: 10.1016/j.yjmcc.2012.04.012
52.
Goodson JM, MacDonald JW, Bammler TK, Chien WM, Chin MT. In utero exposure to diesel exhaust is associated with alterations in neonatal cardiomyocyte transcription, DNA methylation and metabolic perturbation. Particle and Fibre Toxicology. 2019;16:17. doi: 10.1186/s12989-019-0301-9
53.
Chen KH, Cheng ML, Jing YH, Chiu DT, Shiao MS, Chen JK. Resveratrol ameliorates metabolic disorders and muscle wasting in streptozotocin-induced diabetic rats. Am J Physiol Endocrinol Metab. 2011;301:E853–E863. doi: 10.1152/ajpendo.00048.2011
54.
Pinto AR, Ilinykh A, Ivey MJ, Kuwabara JT, D’Antoni ML, Debuque R, Chandran A, Wang L, Arora K, Rosenthal NA, Tallquist MD. Revisiting cardiac cellular composition. Circ Res. 2016;118:400–409. doi: 10.1161/CIRCRESAHA.115.307778
55.
Claycomb WC, Lanson NA, Stallworth BS, Egeland DB, Delcarpio JB, Bahinski A, Izzo NJ. HL-1 cells: a cardiac muscle cell line that contracts and retains phenotypic characteristics of the adult cardiomyocyte. Proc Natl Acad Sci U S A. 1998;95:2979–2984. doi: 10.1073/pnas.95.6.2979

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation Research
Circulation Research
Pages: 6 - 20
PubMed: 35611699

Versions

You are viewing the most recent version of this article.

History

Received: 28 November 2021
Revision received: 28 April 2022
Accepted: 3 May 2022
Published online: 25 May 2022
Published in print: 24 June 2022

Permissions

Request permissions for this article.

Keywords

  1. glycolysis
  2. fibroblasts
  3. integrins
  4. metabolomics
  5. vertebrates

Subjects

Authors

Affiliations

Pei-Chun Chou*
Division of Cardiology, Department of Medicine, Heart Rhythm Center (P.-C.C., C.-M.L., C.-H.W., J.-D.L., Y.-F.H.), Taipei Veterans General Hospital, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Division of Cardiology, Department of Medicine, Heart Rhythm Center (P.-C.C., C.-M.L., C.-H.W., J.-D.L., Y.-F.H.), Taipei Veterans General Hospital, Taiwan.
Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan (C.-M.L., Y.-F.H.).
Ching-Hui Weng
Division of Cardiology, Department of Medicine, Heart Rhythm Center (P.-C.C., C.-M.L., C.-H.W., J.-D.L., Y.-F.H.), Taipei Veterans General Hospital, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Department and Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei (K.-C.Y.).
Mei-Ling Cheng
Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, Taoyuan City, Taiwan (M.-L.C.).
Yuh-Charn Lin
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taiwan (Y.-C.L.).
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Song-Kun Shyue
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Jin-Dian Liu
Division of Cardiology, Department of Medicine, Heart Rhythm Center (P.-C.C., C.-M.L., C.-H.W., J.-D.L., Y.-F.H.), Taipei Veterans General Hospital, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Department of Neurology, Neurological Institute (S.-P.C.), Taipei Veterans General Hospital, Taiwan.
Michael Hsiao
The Genomics Research Center, Academia Sinica, Taipei, Taiwan (M.H.).
Division of Cardiology, Department of Medicine, Heart Rhythm Center (P.-C.C., C.-M.L., C.-H.W., J.-D.L., Y.-F.H.), Taipei Veterans General Hospital, Taiwan.
Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan (P.-C.C., C.-H.W., K.-C.Y., Y.-C.L., R.-B.Y., B.-C.S., S.-K.S., J.-D.L., Y.-F.H.).
Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan (C.-M.L., Y.-F.H.).

Notes

*
P.-C. Chou and C.-M. Liu contributed equally.
Supplemental Material is available at Supplemental Material.
For Sources of Funding and Disclosures, see page 18.
Correspondence to: Yu-Feng Hu, MD, Division of Cardiology, Taipei Veterans General Hospital, 201, Sec. 2, Shih-Pai Road, Taipei, 11217, Taiwan. Email [email protected]

Disclosures

Disclosures None.

Sources of Funding

This work was supported by Taipei Veterans General Hospital (V110C-039, V110B-043, V111C-047, VGHUST111-G6-3-2, VTA111-A-1-2), the Ministry of Science and Technology (110-2628-B-075-015, 110-2314-B-075-063-MY3), National Health Research Institutes (NHRI-109BCC0-MF-202014-02), and Academia Sinica (AS-TM-110-01-01).

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Overexpression of ALDOC Promotes Porcine Intramuscular and Intermuscular Fat Deposition by Activating the AKT-mTORC1 Signaling Pathway, Journal of Agricultural and Food Chemistry, 72, 43, (23893-23907), (2024).https://doi.org/10.1021/acs.jafc.4c07781
    Crossref
  2. Utilizing multi-omics strategies to unravel the molecular basis of heart rhythm, Heart Rhythm, 21, 9, (1761-1762), (2024).https://doi.org/10.1016/j.hrthm.2024.07.005
    Crossref
  3. Can we stop one heart from breaking: triumphs and challenges in cardiac reprogramming, Current Opinion in Genetics & Development, 83, (102116), (2023).https://doi.org/10.1016/j.gde.2023.102116
    Crossref
  4. The role of glycolytic metabolic pathways in cardiovascular disease and potential therapeutic approaches, Basic Research in Cardiology, 118, 1, (2023).https://doi.org/10.1007/s00395-023-01018-w
    Crossref
  5. Harnessing cell reprogramming for cardiac biological pacing, Journal of Biomedical Science, 30, 1, (2023).https://doi.org/10.1186/s12929-023-00970-y
    Crossref
  6. What makes the sinoatrial node tick? A question not for the faint of heart, Philosophical Transactions of the Royal Society B: Biological Sciences, 378, 1879, (2023).https://doi.org/10.1098/rstb.2022.0180
    Crossref
  7. Current approaches for the recreation of cardiac ischaemic environment in vitro, International Journal of Pharmaceutics, 632, (122589), (2023).https://doi.org/10.1016/j.ijpharm.2023.122589
    Crossref
  8. Characterization of diverse populations of sinoatrial node cells and their proliferation potential at single nucleus resolution, Heliyon, 9, 1, (e12708), (2023).https://doi.org/10.1016/j.heliyon.2022.e12708
    Crossref
  9. Novel Insights into the Sinoatrial Node in Single-Cell RNA Sequencing: From Developmental Biology to Physiological Function, Journal of Cardiovascular Development and Disease, 9, 11, (402), (2022).https://doi.org/10.3390/jcdd9110402
    Crossref
  10. Trends in research on sick sinus syndrome: A bibliometric analysis from 2000 to 2022, Frontiers in Cardiovascular Medicine, 9, (2022).https://doi.org/10.3389/fcvm.2022.991503
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this journal for 24 hours

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response