NLRP3 Inflammasome Activation Through Heart-Brain Interaction Initiates Cardiac Inflammation and Hypertrophy During Pressure Overload
Abstract
Background:
Mechanical stress on the heart, such as high blood pressure, initiates inflammation and causes hypertrophic heart disease. However, the regulatory mechanism of inflammation and its role in the stressed heart remain unclear. IL-1β (interleukin-1β) is a proinflammatory cytokine that causes cardiac hypertrophy and heart failure. Here, we show that neural signals activate the NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing 3) inflammasome for IL-1β production to induce adaptive hypertrophy in the stressed heart.
Methods:
C57BL/6 mice, knockout mouse strains for NLRP3 and P2RX7 (P2X purinoceptor 7), and adrenergic neuron-specific knockout mice for SLC17A9, a secretory vesicle protein responsible for the storage and release of ATP, were used for analysis. Pressure overload was induced by transverse aortic constriction. Various animal models were used, including pharmacological treatment with apyrase, lipopolysaccharide, 2′(3′)-O-(4-benzoylbenzoyl)-ATP, MCC950, anti–IL-1β antibodies, clonidine, pseudoephedrine, isoproterenol, and bisoprolol, left stellate ganglionectomy, and ablation of cardiac afferent nerves with capsaicin. Cardiac function and morphology, gene expression, myocardial IL-1β and caspase-1 activity, and extracellular ATP level were assessed. In vitro experiments were performed using primary cardiomyocytes and fibroblasts from rat neonates and human microvascular endothelial cell line. Cell surface area and proliferation were assessed.
Results:
Genetic disruption of NLRP3 resulted in significant loss of IL-1β production, cardiac hypertrophy, and contractile function during pressure overload. A bone marrow transplantation experiment revealed an essential role of NLRP3 in cardiac nonimmune cells in myocardial IL-1β production and cardiac phenotype. Pharmacological depletion of extracellular ATP or genetic disruption of the P2X7 receptor suppressed myocardial NLRP3 inflammasome activity during pressure overload, indicating an important role of ATP/P2X7 axis in cardiac inflammation and hypertrophy. Extracellular ATP induced hypertrophic changes of cardiac cells in an NLRP3- and IL-1β–dependent manner in vitro. Manipulation of the sympathetic nervous system suggested sympathetic efferent nerves as the main source of extracellular ATP. Depletion of ATP release from sympathetic efferent nerves, ablation of cardiac afferent nerves, or a lipophilic β-blocker reduced cardiac extracellular ATP level, and inhibited NLRP3 inflammasome activation, IL-1β production, and adaptive cardiac hypertrophy during pressure overload.
Conclusions:
Cardiac inflammation and hypertrophy are regulated by heart-brain interaction. Controlling neural signals might be important for the treatment of hypertensive heart disease.
Clinical Perspective
What Is New?
•
The nervous system controls cardiac inflammation and hypertrophy during pressure overload through NOD-like receptor pyrin domain-containing protein 3 inflammasome activation.
•
Extracellular ATP released from sympathetic efferent nerve terminals activates the NOD-like receptor pyrin domain-containing protein 3 inflammasome in cardiac nonimmune cells through stimulation of the P2X7 purinergic receptor.
•
Pressure overload is sensed by cardiac afferent nerves to activate sympathetic efferent nerves for ATP release.
What Are the Clinical Implications?
•
Controlling neural signals might have therapeutic potential for the treatment of hypertensive heart disease.
•
Lipophilic β-adrenergic receptor blockers might act on the brain as well as on the heart to inhibit signals of the sympathetic nervous system and cardiac inflammation with its ability to cross the blood-brain barrier.
•
Our mechanism might link psychological distress to heart disease.
Cardiac hypertrophy occurs as an adaptive response to pathological stimuli to maintain cardiac function through reduction in wall stress and energy expenditure.1 However, persistent stress responses lead to contractile dysfunction and heart failure.2 Although inflammation is involved in these processes,3 very little is known about the mechanism that controls cardiac inflammation and hypertrophy.
Inflammation is a complex process in which both immune cells and nonimmune cells are involved. Proinflammatory cytokines, including IL-1β (interleukin-1β), IL-6 (interleukin-6), and TNF-α (tumor necrosis factor-α), induce cellular responses, such as cardiomyocyte hypertrophy and fibroblast and immune cell activation‚ that lead to cardiac hypertrophy and heart failure.3 It has been reported that nonimmune cells, including cardiomyocytes, initiate inflammation by recognizing the endogenous molecules termed danger-associated molecular patterns in the stressed heart, such as heat shock proteins and mitochondrial DNA (mtDNA), through innate immune receptors such as Toll-like receptors (TLRs).4,5 Innate immune receptor signaling activates the transcription factor NF-κB (nuclear factor-κB) for the expression of proinflammatory cytokines, which activate NF-κB again through their receptors in an autocrine and paracrine manner for further inflammatory responses.6 Immune cells such as macrophages and lymphocytes interact with proinflammatory cytokines to modulate their expression pattern.7–9
The NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing 3) inflammasome is a cytosolic multiprotein complex that mediates active IL-1β production.10 IL-1β is a proinflammatory cytokine that is critically involved in the pathophysiology of hypertrophic heart disease.4,11 This complex consists of the Nod-like receptor family protein NLRP3, the ASC (apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain), and procaspase-1. These components are upregulated through the activation of NF-κB, which is called NLRP3 inflammasome priming. On activation of NLRP3 by exogenous pathogens or endogenous danger signals, procaspase-1 is cleaved into 10-kDa and 20-kDa subunits to form the active enzyme, caspase-1. Subsequently, caspase-1 cleaves inactive pro–IL-1β into active IL-1β. Although the NLRP3 inflammasome in immune cells and nonimmune cells has been implicated in cardiovascular stress and disease,9,12–17 how it is activated and contributes to cardiac hypertrophy remain poorly understood.
Here we show that neural signals contribute to NLRP3 inflammasome activation in cardiac nonimmune cells that initiates inflammation and the adaptive programs in the stressed heart through IL-1β production. We find that pressure overload to the left ventricle activates sympathetic efferent nerves (SENs) to secrete extracellular ATP. Extracellular ATP stimulates the purinergic receptor P2X7 (P2X purinoceptor 7) for NLRP3 inflammasome activation in cardiomyocytes, fibroblasts, and vascular endothelial cells. This mechanism is essential for IL-1β production that causes cardiac adaptive hypertrophy in response to mechanical stress. We also demonstrate that cardiac afferent nerve signals contribute to ATP secretion from SEN terminals. These data collectively reveal that cardiac inflammation and hypertrophy are controlled through heart-brain interaction.
Methods
For detailed description of methods, please see the Supplemental Material. The data that support the findings of this study are available from the corresponding author on reasonable request.
Animal Study
All experiments were approved by the University of Tokyo Ethics Committee for Animal Experiments and strictly adhered to the guidelines for animal experiments of the University of Tokyo. Eight- to 12-week-old male mice were used. Wild-type C57BL/6 mice were purchased from Takasugi Experimental Animal Supply. Nlrp3–/– mice were provided by the laboratory of Dr Tschopp (The University of Lausanne, Switzerland).18 P2rx7–/– and Slc17a9flox/flox mice were purchased from the Jackson Laboratory. DBH-Cre mice were provided by RIKEN BRC through the National Bio-Resource Project of MEXT, Japan.19
Pressure overload was induced by transverse aortic constriction (TAC) as described previously.4 Apyrase (4 units, Sigma), lipopolysaccharide (2 mg/kg, InvivoGen), 2′(3′)-O-(4-benzoylbenzoyl)-ATP (BzATP; 5 mg/kg, Sigma), MCC950 (10 mg/kg, Selleck Chemicals), and clonidine (10 μg/kg, Sigma) were injected intraperitoneally daily. Anti–IL-1β antibody or control antibody (100 μg per mouse, R&D Systems) was injected intravenously once. Pseudoephedrine (20 mg·kg–1·d–1, Ieda Chemicals) and bisoprolol hemifumarate (5 mg·kg–1·d–1, Tokyo Chemical Industry) were orally administered to mice by gavage daily. Isoproterenol (Sigma-Aldrich) was administered to mice through an osmotic minipump (30 mg·kg–1·d–1, ALZET mini-osmotic pump, DURECT Corporation). Left stellate ganglionectomy was performed just before TAC or sham operation.20 To ablate primary afferent neurons, subepicardial injection of capsaicin (50 mg/mL, Sigma) dissolved in olive oil (Wako) was performed at 2 weeks before induction of TAC.21
Human Samples
The use of previously obtained human heart biopsy samples from patients with heart failure for daily practice was approved by the Institutional Review Board of the University of Tokyo Hospital, and consent was obtained from all subjects. These samples were fixed in 10% formalin and embedded in paraffin.
Statistical Analyses
Statistical analyses were performed with EZR (Saitama Medical Center, Jichi Medical University, Saitama, Japan), which is a graphical user interface for R (The R Foundation for Statistical Computing).22 Differences in means between 2 groups were analyzed by unpaired 2-tailed t test. More than 2 groups were compared using parametric or nonparametric 1-way ANOVA followed by Holm test or Dunnett test for multiple comparisons. The Kaplan-Meier method with log-rank test was used for survival analysis. Values of P<0.05 were considered statistically significant.
Results
NLRP3 Inflammasome Is Activated in the Stressed Heart
To investigate the involvement of the NLRP3 inflammasome in hypertrophic heart disease, we first examined gene and protein expression of NLRP3 inflammasome components and IL-1β, and caspase-1 activity and cleavage in myocardial tissue. In wild-type mice, NLRP3 mRNA and protein were upregulated during pressure overload caused by TAC (Figure 1A and Figure S1A and S1B). Immunohistochemical staining demonstrated that NLRP3 is heterogeneously expressed in both cardiomyocytes and noncardiomyocytes in the murine pressure-overloaded heart and the human failing heart (Figure 1B). Gene and protein expression levels of other inflammasome components, such as ASC and procaspase-1, and pro–IL-1β were also upregulated in the pressure-overloaded heart, indicating activation of priming signals (Figure S1). Caspase-1 activity and cleaved caspase-1 level were increased during pressure overload and peaked at day 14 after TAC (Figure 1C and Figure S1B). Protein expression of IL-1β was significantly increased during the adaptive hypertrophic phase until day 14 after TAC (Figure 1D). These results indicate that the NLRP3 inflammasome is activated in the stressed heart, especially during the adaptive hypertrophic phase.

Genetic Disruption of NLRP3 Inhibits IL-1β Production and Adaptive Cardiac Hypertrophy
To examine the role of the NLRP3 inflammasome in hypertrophic heart disease, we induced pressure overload by TAC in Nlrp3+/+ and Nlrp3–/– mice.18 We confirmed deletion of NLRP3 protein in hearts from Nlrp3–/– mice by Western blot analysis (Figure S2A). In the absence of pressure overload, cardiac function and morphology did not differ between Nlrp3+/+ and Nlrp3–/– mice (Figure 1E and 1F and Figure S2B). In Nlrp3+/+ mice, pressure overload induced cardiac hypertrophy with preserved contractile function until day 14 (adaptive phase) and heart failure at day 28 after TAC (Figure 1E and Figure S2B). On the other hand, Nlrp3–/– hearts showed attenuated cardiac hypertrophy, but greater left ventricular dilation with impaired contractile function compared with Nlrp3+/+ hearts in the adaptive phase after TAC (Figure 1E and Figure S2B). In the heart-failure phase, cardiac function and chamber size were similar between Nlrp3+/+ and Nlrp3–/– hearts, whereas cardiac hypertrophy remained attenuated in Nlrp3–/– hearts (Figure 1E). Histological assessment demonstrated significantly smaller cardiomyocytes and reduced cardiac fibrosis and macrophage infiltration in Nlrp3–/– hearts compared with Nlrp3+/+ hearts during pressure overload (Figure 1F and Figure S2C–S2E). In the adaptive phase, angiogenesis was suppressed in Nlrp3–/– hearts (Figure 1F and Figure S2F). Mortality after TAC was higher in Nlrp3–/– mice than in Nlrp3+/+ mice (Figure 1G), whereas all sham-operated Nlrp3+/+ and Nlrp3–/– mice survived (n=8 for each). All deaths of Nlrp3–/– mice were observed in the adaptive phase. Hemodynamic measurement revealed higher left ventricle end-diastolic pressure and smaller absolute values of maximum and minimum dp/dt in Nlrp3–/– mice than in Nlrp3+/+ mice after 14 days of pressure overload, whereas blood pressure in the ascending aorta was similar between these mice (Figure 1H). There were no significant differences in hemodynamic parameters between Nlrp3+/+ and Nlrp3–/– mice at 28 days after TAC or sham operation (Figure S2G). These results collectively indicate that genetic disruption of NLRP3 prevented pathological cardiac remodeling but might have impaired cardiac adaptative response to pressure overload.
At 14 days after TAC, the mRNA levels of hypertrophic marker genes (Nppa and Myh7), a fibrosis-related gene (Col1a1), an angiogenesis-related gene (Vegfa), and inflammation-related genes (Il6, Mcp1, and Tnfa) were consistently upregulated in wild-type hearts, whereas they were suppressed in Nlrp3–/– hearts (Figure 2A). Caspase-1 activity and cleaved caspase-1 and IL-1β levels were lower in Nlrp3–/– hearts than in wild-type hearts (Figure 2B–2D). Mitochondrial dysfunction is one of the characteristics of failing cardiomyocytes. To examine mitochondrial function, we assessed mtDNA content and oxidative damage and expression levels of genes associated with mitochondrial oxidative phosphorylation in the heart (Figure 2E and Figure S3).23 Although Nlrp3+/+ failing hearts showed the decrease in mtDNA content and expression levels of some of the oxidative phosphorylation genes and the increase in mtDNA oxidative damage, no significant changes in these indicators of mitochondrial function were observed in Nlrp3–/– hearts during pressure overload. These results indicate that mitochondrial function during pressure overload was not impaired in Nlrp3–/– hearts. Thus, contractile dysfunction in Nlrp3–/– mice during pressure overload might not be attributable to pathological changes of cardiomyocytes, but to insufficient hemodynamic adaptation to pressure overload.

To dissect the role of NLRP3 in immune cells from that in nonimmune cells, we performed a bone marrow transplantation experiment (Figures 2F and 2G and Figure S4). Caspase-1 activation and IL-1β production in the heart did not differ between wild-type mice transplanted with Nlrp3+/+ and Nlrp3–/– bone marrow during pressure overload (Figure 2F and 2G and Figure S4A). Both mice showed cardiac hypertrophy with preserved contractile function after 14 days of TAC to a similar extent (Figure S4B and S4C). Cardiomyocyte hypertrophy, fibrosis, macrophage infiltration, and angiogenesis in pressure-overloaded hearts were consistently similar between wild-type mice with Nlrp3+/+ and Nlrp3–/– bone marrow (Figure S4D–S4G). No significant differences in hemodynamic parameters were observed between these mice (Figure S4H). These data collectively indicate that NLRP3 inflammasome activation in cardiac nonimmune cells initiate inflammation and adaptive cardiac hypertrophy in response to pressure overload.
ATP/P2X7 Axis Contributes to NLRP3 Inflammasome Activation and Cardiac Hypertrophy During Pressure Overload
Three models of NLRP3 inflammasome activation have been widely suggested: potassium efflux through the P2X7 purinergic receptor stimulated by extracellular ATP, reactive oxygen species–dependent dissociation of TXNIP (thioredoxin-interacting protein) from thioredoxin, and cathepsin B release through lysosomal rupture attributable to mechanical insult by crystalline ligands.24 We focused on the first mechanism because, during pressure overload, TXNIP level was not upregulated in the heart (Figure S5A) and crystalline ligands are considered not to be produced.
To investigate the involvement of extracellular ATP and the P2X7 receptor in NLRP3 inflammasome activation and cardiac hypertrophy, we induced pressure overload in wild-type mice treated with an ATP diphosphohydrolase, apyrase,25 and P2rx7–/– mice. In these mice, caspase-1 activation and IL-1β production were suppressed during pressure overload compared with wild-type mice treated with vehicle or P2rx7+/+ mice (Figure 3A–3D and Figure S5B and S5C). We also found attenuated cardiac hypertrophy, greater ventricular dilation and contractile dysfunction with smaller cardiomyocytes, reduced cardiac fibrosis and macrophage infiltration, and lower capillary density in these mice compared with control mice on day 14 after TAC (Figure 3E and 3F and Figures S5D, S5E, and S6A–S6H). Hemodynamic measurement showed that apyrase treatment or genetic disruption of the P2X7 receptor led to higher left ventricle end-diastolic pressure and smaller maximum dp/dt in pressure-overloaded hearts without affecting blood pressure in the ascending aorta (Figure S6I and S6J). These data indicate that extracellular ATP and the P2X7 receptor are required for NLRP3 inflammasome activation and adaptive cardiac hypertrophy in response to pressure overload.

Role of ATP/P2X7 Axis in the Heart
To clarify the role of the ATP/P2X7 axis and the NLRP3 inflammasome in cardiac cells, we assessed their effect on cardiomyocyte hypertrophy and proliferation of fibroblasts and vascular endothelial cells, which are important processes in cardiac hypertrophy.2,4 We previously showed that TLR2 signaling is essential for IL-1β mRNA production in the stressed heart.4 A specific ligand for TLR2, Pam3CSK4,4 induced cardiomyocyte hypertrophy and proliferation of fibroblasts and vascular endothelial cells (Figure 3G–3I). Treatment with ATP in combination with Pam3CSK4 resulted in further hypertrophy and proliferation. Pharmacological inhibition of the P2X7 receptor, Nlrp3 knockdown, or anti–IL-1β neutralization antibody treatment inhibited the synergistic effects of ATP and Pam3CSK4 on cardiomyocytes, fibroblasts, and vascular endothelial cells (Figure 3G–3I and Figure S7). These data indicate that the ATP/P2X7 axis contributes to hypertrophic responses of cardiac cells through the NLRP3 inflammasome activation and IL-1β production.
We next examined whether activation of the P2X7 receptor, with or without an inflammasome priming signal, induces cardiac hypertrophy in vivo. Wild-type mice treated with a combination of BzATP, a P2X7 receptor agonist, and lipopolysaccharide, a ligand of TLR4, for 14 days showed cardiac hypertrophy and increased myocardial caspase-1 activity and IL-1β level with preserved systolic function and chamber size, compared with those treated with vehicle (Figure S8A–S8C). Histological assessment demonstrated cardiomyocyte hypertrophy and augmented interstitial fibrosis in wild-type mice treated with the combination (Figure S8D–S8F), which was confirmed by the increase in expression levels of Myh7 and Col1a1(Figure S8G–S8I). At our dose, BzATP or lipopolysaccharide alone did not induce cardiac hypertrophy (Figure S8A). Treatment with BzATP alone increased myocardial caspase-1 activity, but to a lesser extent than the combination treatment, which did not lead to IL-1β production (Figure S8B and S8C). Treatment with MCC950, a potent and specific NLRP3 inhibitor, or anti-IL-1β antibodies suppressed caspase-1 activity, IL-1β level, and cardiac hypertrophy without affecting systolic function or chamber size in wild-type mice treated with the combination of BzATP and lipopolysaccharide, compared with control treatment (Figure S8A–S8I). Blood pressure was not affected by these treatments (Figure S8J). The P2X7 receptor has been implicated in cardiomyocyte hypertrophy and cellular survival through NLRP3 inflammasome-independent mechanisms.26 Our data suggest that P2X7 receptor signaling might regulate cardiac hypertrophic changes in vivo mainly through the NLRP3 inflammasome activation and IL-1β production, rather than through NLRP3 inflammasome-independent mechanisms. In addition, IL-1β might affect myocardial caspase-1 activity in a positive-feedback manner through NF-κB activation.
SENs Release ATP for NLRP3 Inflammasome Activation
To examine the dynamics of extracellular ATP during pressure overload, we measured its concentration in the heart with an ATP-sensing electrode (Figure 4A and Figure S9A).27 We found that extracellular ATP was increased in wild-type hearts on day 14 after TAC compared with sham-operated mice, whereas apyrase treatment reduced its concentration (Figure 4B and Figure S9B–S9D). We also visualized extracellular ATP in vivo by expressing the engineered firefly luciferase, called pmeLUC, which localizes to the outer aspect of the plasma membrane with the catalytic site facing the extracellular environment,28 in cardiomyocytes through the use of adeno-associated virus, and confirmed the increase of extracellular ATP by pressure overload (Figures 4C–4F). As a danger signal, ATP is released by necrotic or apoptotic cells in damaged organs. In the pressure-overloaded heart, however, massive cell death does not occur; thus, extracellular ATP may be released by active transport from living cells. It is reported that cardiac cells can release ATP only below a physiologically active concentration.29 Because ATP is a neurotransmitter and is secreted from nerve terminals,30 we hypothesized that the autonomic nervous system may be the main source of extracellular ATP for NLRP3 inflammasome activation in the heart during pressure overload.

We first examined the role of the sympathetic nervous system (SNS), which innervates the left ventricle more abundantly than does the parasympathetic nervous system, in ATP release and NLRP3 inflammasome activation in the pressure-overloaded heart.31 Immunohistochemistry confirmed the presence of catecholaminergic nerve fibers in the epicardial nerve bundles around the left ventricle (Figure 4G). No significant difference in total areas of epicardial catecholaminergic nerve fibers was observed between sham-operated and TAC-operated hearts at day 14 after the operations. We measured extracellular ATP level in the left ventricle of wild-type mice treated with ablation of the left stellate ganglion,20 the main source of the SEN terminals in the left ventricle, and clonidine, an SNS suppressant through stimulation of the α2-adrenergic receptor in the central nervous system. In these mice, extracellular ATP level during pressure overload was suppressed compared with wild-type mice without any treatment (Figure 4B and Figure S9E and S9F). In addition, these mice showed impairment of the cardiac adaptive response to pressure overload, with suppressed caspase-1 activation and IL-1β production, whereas systolic blood pressure was not affected by ablation of the left stellate ganglionectomy or clonidine treatment (Figure 4H–4K and Figure S10 and S11). These data indicate that SEN signals to the heart are required for the increase of extracellular ATP level and NLRP3 inflammasome activation to induce cardiac adaptive hypertrophy during pressure overload. In addition, it is suggested that the central nervous system is involved in this mechanism.
Next, we investigated whether SNS activation is sufficient for the increase of extracellular ATP level and NLRP3 inflammasome activation in the heart. Sympathetic activation by pseudoephedrine increased extracellular ATP level and caspase-1 activity in wild-type hearts (Figure 4B and 4L and Figure S9G and S12A). In pseudoephedrine-treated Nlrp3–/– hearts, caspase-1 activation was suppressed compared with that in pseudoephedrine-treated wild-type hearts (Figure 4L and Figure S12A). We did not find significant differences in systolic blood pressure and heart rate between pseudoephedrine-treated Nlrp3+/+ and Nlrp3–/– mice, although pseudoephedrine slightly increased systolic blood pressure in these mice compared with vehicle (Figure S12B and S12C). Thus, sympathetic activation induces extracellular ATP release and NLRP3 inflammasome-dependent caspase-1 activation in the heart.
At our dose, pseudoephedrine did not induce IL-1β production and cardiac hypertrophy in both wild-type and Nlrp3–/– mice, although pseudoephedrine slightly increased macrophage infiltration (Figure 4L and Figure S12D–S12I). These findings suggest that NLRP3 inflammasome activation by the SNS alone might not be sufficient for IL-1β production. Other signaling pathways such as TLR signaling, which upregulates IL-1β mRNA, might be necessary for IL-1β production and cardiac hypertrophy.
We next investigated the effect of adrenergic signals on extracellular ATP level and NLRP3 inflammasome activation in the heart. Infusion of isoproterenol, a β-adrenergic receptor agonist, induced cardiac hypertrophy with fibrosis, macrophage infiltration, and angiogenesis in both wild-type and Nlrp3–/– mice (Figure 4M and Figure S13A–S13F). No significant differences in systolic blood pressure and heart rate were observed between isoproterenol-treated Nlrp3+/+ and Nlrp3–/– mice, although isoproterenol slightly increased systolic blood pressure and heart rate compared with vehicle (Figure S13G and S13H). Norepinephrine, a main neurotransmitter of the SENs, induced cardiomyocyte hypertrophy and proliferation of cardiac fibroblasts and vascular endothelial cells in an NLRP3 inflammasome-independent manner in vitro (Figure S14). Isoproterenol did not significantly increase extracellular ATP level in the heart (Figure 4B and Figure S9H). Caspase-1 activity and cleaved caspase-1 level were increased in both isoproterenol-treated wild-type and Nlrp3–/– mice (Figure 4M and Figure S13I). IL-1β level was not increased in these mice (Figure 4M).These data collectively suggest that, in the pressure-overloaded heart, ATP might be released mainly from SEN terminals rather than from cardiac cells, stimulated by adrenergic signals. In addition, adrenergic signals might be able to induce caspase-1 activation and cardiac hypertrophy in an NLRP3 inflammasome-independent manner.
ATP released from the sympathetic nerve terminals has been reported to modulate presynaptic norepinephrine release through activation of purinergic receptors.32,33 To examine the effect of extracellular ATP on presynaptic norepinephrine release and the role of norepinephrine on NLRP3 inflammasome activation and cardiac phenotype in the pressure-overloaded heart, we next measured myocardial and plasma norepinephrine levels. We found that pressure overload by TAC increases myocardial and plasma norepinephrine levels in wild-type mice (Figure S15). No significant differences in myocardial and plasma norepinephrine levels were detected between TAC-operated Nlrp3+/+ and Nlrp3–/– mice. Apyrase treatment or genetic disruption of the P2X7 receptor did not affect norepinephrine level in the heart or plasma on day 14 after TAC or sham operation. We did not observe the increase of myocardial norepinephrine level after TAC in wild-type mice treated with ablation of the left stellate ganglion, whereas this treatment did not have an effect on plasma norepinephrine level, which might reflect the contribution of the left stellate ganglion to sympathetic innervation in the left ventricle. These data suggest that ATP might regulate presynaptic norepinephrine release with various positive and negative feedbacks through purinergic receptors, including P2X and P2Y receptors,32 which might explain why myocardial norepinephrine level was not altered by ATP depletion or genetic disruption of the P2X7 receptor. In addition, our data indicate that norepinephrine might not contribute to NLRP3 inflammasome activation and cardiac phenotype in our TAC model. Furthermore, pressure overload might increase systemic sympathetic neural activity and norepinephrine in the blood, whereas the increase of norepinephrine in the pressure-overloaded heart might be attributable mainly to its release from the SEN terminals in the heart.
ATP From SENs Is Essential for Cardiac Hypertrophy
Vesicular nucleotide transporter (also known as Slc17a9) is a secretory vesicle protein that is responsible for the storage and release of ATP in neurons.34 To clearly dissect the role of ATP release from that of norepinephrine release by the SNS in the pressure-overloaded heart, we crossed mice bearing an Slc17a9flox allele with transgenic mice expressing Cre recombinase under the control of the dopamine β-hydroxylase (DBH) promoter to generate Slc17a9flox/flox;DBH-Cre+ (Slc17a9–/–) mice.19 The DBH promoter drives gene expression in noradrenergic and adrenergic cell groups, including postganglionic neurons in the SNS (Figure 5A). In Slc17a9–/– mice, ATP release from SEN terminals is inhibited, whereas norepinephrine release remains intact. We used Slc17a9flox/flox;DBH-Cre- (Slc17a9+/+) littermates as controls. Slc17a9–/– mice did not display a cardiac structural or functional deficit at baseline. In Slc17a9–/– mice, extracellular ATP release, caspase-1 activation, and IL-1β production in the heart were suppressed compared with those in Slc17a9+/+ mice on day 14 after TAC (Figures 4B and 5B–5D and Figure S9I and S9J). Myocardial and plasma norepinephrine levels were comparable between Slc17a9+/+ and Slc17a9–/– mice (Figure 5E and 5F). Slc17a9–/– mice showed attenuated cardiac hypertrophy and contractile dysfunction, with reduced cardiomyocyte hypertrophy, fibrosis, capillary density, and macrophage infiltration compared with Slc17a9+/+ mice, indicating impairment of the adaptive mechanisms in response to pressure overload (Figure 5G–5L). Higher left ventricle end-diastolic pressure and smaller maximum dp/dt in pressure-overloaded hearts were consistently observed in Slc17a9–/– mice compared with Slc17a9+/+ mice, although blood pressure in the ascending aorta was comparable between these mice (Figure 5M). Collectively, SEN signals directly regulate NLRP3 inflammasome activation and cardiac adaptive hypertrophy through ATP release.

Role of Cardiac Afferent Nerves in ATP Release From SENs and NLRP3 Inflammasome Activation
We next assessed whether afferent input signals from the heart contribute to ATP release from SENs and NLRP3 inflammasome activation. No significant difference in total areas of epicardial primary afferent nerve fibers was observed between sham-operated and TAC-operated hearts at day 14 after the operations (Figure 6A). We treated the heart with capsaicin, an agonist for the transient receptor potential vanilloid type 1 channel that is predominantly expressed on the terminals of primary sensory neurons, to ablate afferent nerves from the heart.8,21 Immunohistochemical staining confirmed ablation of primary sensory nerve fibers without depletion of catecholaminergic nerve fibers consistently with the previous reports (Figure 6B). Cardiac function and morphology did not differ between sham-operated wild-type mice treated with capsaicin or vehicle (Figures 6C–6H). Capsaicin-treated hearts showed reduced extracellular ATP level and suppressed caspase-1 activation and IL-1β production during pressure overload compared with vehicle-treated mice (Figures 4B and 6I–6K and Figure S9K). Ablation of cardiac afferent nerves attenuated cardiac hypertrophy but resulted in greater left ventricular dilation and impaired systolic function with higher left ventricle end-diastolic pressure and smaller absolute values of maximum and minimum dp/dt on day 14 after TAC, but this treatment did not affect systolic blood pressure in the ascending aorta (Figure 6C, 6D, and 6L). Histological assessment revealed reduced cardiomyocyte hypertrophy, interstitial fibrosis, capillary density, and macrophage infiltration by capsaicin treatment during pressure overload (Figure 6E–6H). Thus, ablation of cardiac afferent nerves impaired adaptive cardiac hypertrophy. These data indicate that afferent nerve signals are required for ATP release from SEN terminals and NLRP3 inflammasome activation for cardiac adaptive hypertrophy during pressure overload.

NLRP3 Inflammasome Activity in Isolated Perfused Hearts Ex Vivo
To further examine the significance of neural signals in NLRP3 inflammasome activation in the pressure-overloaded heart, we performed an ex vivo experiment using the Langendorff perfused heart model.35 In this experimental model, input and output neural signals in the heart are completely ablated. We found no significant differences in caspase-1 activity and IL-1β production between hearts with and without a pressure overload of ≈40 mm Hg for 60 minutes (Figure S16). Our data suggest that pressure overload itself might not be sufficient for NLRP3 inflammasome activation and IL-1β production, although our experiment could assess only short-term responses.
Bisoprolol Inhibits Extracellular ATP Release and NLRP3 Inflammasome Activation
Lipophilic β-adrenergic receptor blockers are used as standard therapy for cardiac remodeling and heart failure.36 These drugs can cross the blood-brain barrier. To investigate the effect of lipophilic β-adrenergic receptor blockers on extracellular ATP release and NLRP3 inflammasome activation in the heart, we induced pressure overload in wild-type mice treated with a lipophilic β-adrenergic receptor blocker, bisoprolol, or vehicle. Our tested dose of bisoprolol lowered heart rate, but it did not change systolic blood pressure in wild-type mice (Figure 7A). We assessed extracellular ATP levels by using the pmeLUC system and found that bisoprolol reduced extracellular ATP in TAC-operated hearts (Figure 7B and 7C). In bisoprolol-treated mice, caspase-1 activation and IL-1β production were suppressed during pressure overload compared with vehicle-treated mice (Figure 7D and 7E). We observed attenuated cardiac hypertrophy, greater ventricular dilation, and reduced ejection fraction with smaller cardiomyocytes, reduced cardiac fibrosis and macrophage infiltration, and lower capillary density in bisoprolol-treated mice compared with control mice at day 14 after TAC (Figure 7F–7K). Ejection fraction remained >50% in bisoprolol-treated mice. These results indicate that bisoprolol ameliorated cardiac inflammation and pathological cardiac remodeling, possibly in part, by inhibiting extracellular ATP release from the sympathetic nerve terminals, although our tested dose of bisoprolol suppressed systolic function under persistent pressure overload.

Discussion
In this study, we demonstrated that neural signals control cardiac inflammation and hypertrophy through NLRP3 inflammasome activation and IL-1β production during pressure overload (Figure 8). IL-1β is a key proinflammatory cytokine that contributes to the pathophysiology of hypertrophic heart disease.4,11 Active IL-1β production is tightly controlled in a 2-step process.10 The first step upregulates the inactive precursor, pro–IL-1β, by promoting the transcription of IL-1β gene. The second step processes pro–IL-1β into active IL-1β by activating caspase-1. In the heart, the first step is activated by local proinflammatory mechanisms, in which danger-associated molecular patterns from damaged cardiac cells stimulate innate immune receptors for NF-κB activation in an autocrine and paracrine manner.4,5 Our data show that the second step is regulated by the nervous system, including the central nervous system, through NLRP3 inflammasome activation. Thus, both local mechanisms and organ communications are necessary for proinflammatory process in the heart. Together with the previous reports that showed the involvement of the central nervous system in the pathophysiology of heart disease,8,21,37 our mechanism might link psychological distress to heart disease.38

Inflammation is a double-edged sword that has both protective and harmful effects under various pathological conditions.39,40 In our study, genetic disruption of NLRP3 resulted in contractile dysfunction and hemodynamic maladaptation with high mortality after TAC, whereas it inhibited cardiac hypertrophy and progression of pathological cardiac remodeling during pressure overload. NLRP3 inflammasome inhibitors have recently been suggested to be effective for the treatment of cardiovascular disease.12,15,41,42 Our findings suggest that, similarly to β-adrenergic receptor blockers, careful titration of NLRP3 inflammasome inhibitors might be necessary for inhibition of pathological cardiac remodeling without hemodynamic maladaptation.
Our results suggest that ATP, rather than norepinephrine, might be the main neurotransmitter that initiates cardiac inflammation and hypertrophy, at least in the adaptive phase of pressure overload, although β-adrenergic signals in the heart might have an interaction with the NLRP3 inflammasome and contribute to pathological cardiac remodeling in both NLRP3 inflammasome-dependent and -independent manners. Taken together with the data on bisoprolol-treated mice, lipophilic β-adrenergic receptor blockers, which are used as standard therapy for cardiac remodeling and heart failure,36 might act on the brain as well as on the heart to inhibit SNS signals and cardiac inflammation with its ability to cross the blood-brain barrier. Further studies are needed to clarify the crosstalk between NLRP3 inflammasome activation and β-adrenergic signals in hypertensive heart disease.
In conclusion, NLRP3 inflammasome activation through heart-brain interaction initiates cardiac inflammation and hypertrophy during pressure overload. The nervous system could be a therapeutic target for the treatment of hypertensive heart disease, although fine-tuning might be necessary.
For a comprehensive discussion of the study, see the Expanded Discussion in the Supplemental Material.
Article Information
Supplemental Material
Expanded Methods
Expanded Discussion
Figures S1–S16
Table S1
References 43–50
Acknowledgments
The authors are grateful to Dr Tschopp (The University of Lausanne, Switzerland) and Dr Di Virgilio (The University of Ferrara, Italy) for providing Nlrp3–/– mice and the plasmid harboring pmeLUC, respectively. Drs Higashikuni and Sata conceived the study. Dr Higashikuni designed and performed experiments and analyzed data. Drs Liu and Y. Tanaka assisted with animal experiments and data analysis. Drs Numata and Takimoto established and performed the Langendorff heart experiment. Drs K. Tanaka, Fukuda, and Hirata assisted with histological analysis and discussed analyses and results of all experiments. Dr Imamura assisted with the analysis of human samples. Drs Komuro and Sata supervised the research and provided scientific guidance and analysis. Drs Higashikuni and Sata wrote the manuscript with input from all authors. All authors approved the article.
Footnote
Nonstandard Abbreviations and Acronyms
- ASC
- apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain
- IL
- interleukin
- mtDNA
- mitochondrial DNA
- NF-κB
- nuclear factor-κB
- NLRP3
- nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing 3
- SEN
- sympathetic efferent nerve
- TAC
- transverse aortic constriction
- TLR
- Toll-like receptor
- TNF-α
- tumor necrosis factor-α
- TXNIP
- thioredoxin-interacting protein
Supplemental Material
REFERENCES
1.
Frey N, Olson EN. Cardiac hypertrophy: the good, the bad, and the ugly. Annu Rev Physiol. 2003;65:45–79. doi: 10.1146/annurev.physiol.65.092101.142243
2.
Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol. 2006;7:589–600. doi: 10.1038/nrm1983
3.
Mann DL. Innate immunity and the failing heart: the cytokine hypothesis revisited. Circ Res. 2015;116:1254–1268. doi: 10.1161/CIRCRESAHA.116.302317
4.
Higashikuni Y, Tanaka K, Kato M, Nureki O, Hirata Y, Nagai R, Komuro I, Sata M. Toll-like receptor-2 mediates adaptive cardiac hypertrophy in response to pressure overload through interleukin-1β upregulation via nuclear factor κB activation. J Am Heart Assoc. 2013;2:e000267. doi: 10.1161/JAHA.113.000267
5.
Oka T, Hikoso S, Yamaguchi O, Taneike M, Takeda T, Tamai T, Oyabu J, Murakawa T, Nakayama H, Nishida K, et al. Mitochondrial DNA that escapes from autophagy causes inflammation and heart failure. Nature. 2012;485:251–255. doi: 10.1038/nature10992
6.
Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010;11:373–384. doi: 10.1038/ni.1863
7.
Epelman S, Liu PP, Mann DL. Role of innate and adaptive immune mechanisms in cardiac injury and repair. Nat Rev Immunol. 2015;15:117–129. doi: 10.1038/nri3800
8.
Fujiu K, Shibata M, Nakayama Y, Ogata F, Matsumoto S, Noshita K, Iwami S, Nakae S, Komuro I, Nagai R, et al. A heart-brain-kidney network controls adaptation to cardiac stress through tissue macrophage activation. Nat Med. 2017;23:611–622. doi: 10.1038/nm.4326
9.
Sano S, Oshima K, Wang Y, MacLauchlan S, Katanasaka Y, Sano M, Zuriaga MA, Yoshiyama M, Goukassian D, Cooper MA, et al. Tet2-mediated clonal hematopoiesis accelerates heart failure through a mechanism involving the IL-1beta/NLRP3 inflammasome. J Am Coll Cardiol. 2018;71:875–886. doi: 10.1016/j.jacc.2017.12.037
10.
Franchi L, Eigenbrod T, Munoz-Planillo R, Nunez G. The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol. 2009;10:241–247. doi: 10.1038/ni.1703
11.
Honsho S, Nishikawa S, Amano K, Zen K, Adachi Y, Kishita E, Matsui A, Katsume A, Yamaguchi S, Nishikawa K, et al. Pressure-mediated hypertrophy and mechanical stretch induces IL-1 release and subsequent IGF-1 generation to maintain compensative hypertrophy by affecting Akt and JNK pathways. Circ Res. 2009;105:1149–1158. doi: 10.1161/CIRCRESAHA.109.208199
12.
Fuster JJ, MacLauchlan S, Zuriaga MA, Polackal MN, Ostriker AC, Chakraborty R, Wu CL, Sano S, Muralidharan S, Rius C, et al. Clonal hematopoiesis associated with TET2 deficiency accelerates atherosclerosis development in mice. Science. 2017;355:842–847. doi: 10.1126/science.aag1381
13.
Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 2011;123:594–604. doi: 10.1161/CIRCULATIONAHA.110.982777
14.
Mezzaroma E, Toldo S, Farkas D, Seropian IM, Van Tassell BW, Salloum FN, Kannan HR, Menna AC, Voelkel NF, Abbate A. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc Natl Acad Sci U S A. 2011;108:19725–19730. doi: 10.1073/pnas.1108586108
15.
Suetomi T, Willeford A, Brand CS, Cho Y, Ross RS, Miyamoto S, Brown JH. Inflammation and NLRP3 inflammasome activation initiated in response to pressure overload by Ca2+/calmodulin-dependent protein kinase II δ signaling in cardiomyocytes are essential for adverse cardiac remodeling. Circulation. 2018;138:2530–2544. doi: 10.1161/CIRCULATIONAHA.118.034621
16.
Xiao H, Li H, Wang JJ, Zhang JS, Shen J, An XB, Zhang CC, Wu JM, Song Y, Wang XY, et al. IL-18 cleavage triggers cardiac inflammation and fibrosis upon beta-adrenergic insult. Eur Heart J. 2018;39:60–69. doi: 10.1093/eurheartj/ehx261
17.
Wang Y, Wu Y, Chen J, Zhao S, Li H. Pirfenidone attenuates cardiac fibrosis in a mouse model of TAC-induced left ventricular remodeling by suppressing NLRP3 inflammasome formation. Cardiology. 2013;126:1–11. doi: 10.1159/000351179
18.
Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440:237–241. doi: 10.1038/nature04516
19.
Matsushita N, Kobayashi K, Miyazaki J, Kobayashi K. Fate of transient catecholaminergic cell types revealed by site-specific recombination in transgenic mice. J Neurosci Res. 2004;78:7–15. doi: 10.1002/jnr.20229
20.
Shimizu I, Yoshida Y, Katsuno T, Tateno K, Okada S, Moriya J, Yokoyama M, Nojima A, Ito T, Zechner R, et al. p53-induced adipose tissue inflammation is critically involved in the development of insulin resistance in heart failure. Cell Metab. 2012;15:51–64. doi: 10.1016/j.cmet.2011.12.006
21.
Okada S, Yokoyama M, Toko H, Tateno K, Moriya J, Shimizu I, Nojima A, Ito T, Yoshida Y, Kobayashi Y, et al. Brain-derived neurotrophic factor protects against cardiac dysfunction after myocardial infarction via a central nervous system-mediated pathway. Arterioscler Thromb Vasc Biol. 2012;32:1902–1909. doi: 10.1161/ATVBAHA.112.248930
22.
Kanda Y. Investigation of the freely available easy-to-use software ‘EZR’ for medical statistics. Bone Marrow Transplant. 2013;48:452–458. doi: 10.1038/bmt.2012.244
23.
Sato M, Kadomatsu T, Miyata K, Warren JS, Tian Z, Zhu S, Horiguchi H, Makaju A, Bakhtina A, Morinaga J, et al. The lncRNA Caren antagonizes heart failure by inactivating DNA damage response and activating mitochondrial biogenesis. Nat Commun. 2021;12:2529. doi: 10.1038/s41467-021-22735-7
24.
Guo H, Callaway JB, Ting JP. Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21:677–687. doi: 10.1038/nm.3893
25.
Wilhelm K, Ganesan J, Muller T, Durr C, Grimm M, Beilhack A, Krempl CD, Sorichter S, Gerlach UV, Juttner E, et al. Graft-versus-host disease is enhanced by extracellular ATP activating P2X7R. Nat Med. 2010;16:1434–1438. doi: 10.1038/nm.2242
26.
Shokoples BG, Paradis P, Schiffrin EL. P2X7 receptors: an untapped target for the management of cardiovascular disease. Arterioscler Thromb Vasc Biol. 2021;41:186–199. doi: 10.1161/ATVBAHA.120.315116
27.
Gourine AV, Llaudet E, Dale N, Spyer KM. ATP is a mediator of chemosensory transduction in the central nervous system. Nature. 2005;436:108–111. doi: 10.1038/nature03690
28.
Pellegatti P, Falzoni S, Pinton P, Rizzuto R, Di Virgilio F. A novel recombinant plasma membrane-targeted luciferase reveals a new pathway for ATP secretion. Mol Biol Cell. 2005;16:3659–3665. doi: 10.1091/mbc.e05-03-0222
29.
Nishida M, Sato Y, Uemura A, Narita Y, Tozaki-Saitoh H, Nakaya M, Ide T, Suzuki K, Inoue K, Nagao T, et al. P2Y6 receptor-Galpha12/13 signalling in cardiomyocytes triggers pressure overload-induced cardiac fibrosis. EMBO J. 2008;27:3104–3115. doi: 10.1038/emboj.2008.237
30.
Burnstock G. Historical review: ATP as a neurotransmitter. Trends Pharmacol Sci. 2006;27:166–176. doi: 10.1016/j.tips.2006.01.005
31.
Kanazawa H, Ieda M, Kimura K, Arai T, Kawaguchi-Manabe H, Matsuhashi T, Endo J, Sano M, Kawakami T, Kimura T, et al. Heart failure causes cholinergic transdifferentiation of cardiac sympathetic nerves via gp130-signaling cytokines in rodents. J Clin Invest. 2010;120:408–421. doi: 10.1172/JCI39778
32.
Sperlagh B, Heinrich A, Csolle C. P2 receptor-mediated modulation of neurotransmitter release-an update. Purinergic Signal. 2007;3:269–284. doi: 10.1007/s11302-007-9080-0
33.
Li J, King NC, Sinoway LI. Interstitial ATP and norepinephrine concentrations in active muscle. Circulation. 2005;111:2748–2751. doi: 10.1161/CIRCULATIONAHA.104.510669
34.
Larsson M, Sawada K, Morland C, Hiasa M, Ormel L, Moriyama Y, Gundersen V. Functional and anatomical identification of a vesicular transporter mediating neuronal ATP release. Cereb Cortex. 2012;22:1203–1214. doi: 10.1093/cercor/bhr203
35.
Takimoto E, Champion HC, Belardi D, Moslehi J, Mongillo M, Mergia E, Montrose DC, Isoda T, Aufiero K, Zaccolo M, et al. cGMP catabolism by phosphodiesterase 5A regulates cardiac adrenergic stimulation by NOS3-dependent mechanism. Circ Res. 2005;96:100–109. doi: 10.1161/01.RES.0000152262.22968.72
36.
Bristow MR. β-Adrenergic receptor blockade in chronic heart failure. Circulation. 2000;101:558–569. doi: 10.1161/01.cir.101.5.558
37.
Leenen FH. Brain mechanisms contributing to sympathetic hyperactivity and heart failure. Circ Res. 2007;101:221–223. doi: 10.1161/CIRCRESAHA.107.158261
38.
Jiang W, Kuchibhatla M, Cuffe MS, Christopher EJ, Alexander JD, Clary GL, Blazing MA, Gaulden LH, Califf RM, Krishnan RR, et al. Prognostic value of anxiety and depression in patients with chronic heart failure. Circulation. 2004;110:3452–3456. doi: 10.1161/01.CIR.0000148138.25157.F9
39.
Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, et al; CANTOS Trial Group. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377:1119–1131. doi: 10.1056/NEJMoa1707914
40.
Gomez D, Baylis RA, Durgin BG, Newman AAC, Alencar GF, Mahan S, St Hilaire C, Muller W, Waisman A, Francis SE, et al. Interleukin-1β has atheroprotective effects in advanced atherosclerotic lesions of mice. Nat Med. 2018;24:1418–1429. doi: 10.1038/s41591-018-0124-5
41.
Zahid A, Li B, Kombe AJK, Jin T, Tao J. Pharmacological Inhibitors of the NLRP3 Inflammasome. Front Immunol. 2019;10:2538. doi: 10.3389/fimmu.2019.02538
42.
Yao C, Veleva T, Scott L, Cao S, Li L, Chen G, Jeyabal P, Pan X, Alsina KM, Abu-Taha ID, et al. Enhanced cardiomyocyte NLRP3 inflammasome signaling promotes atrial fibrillation. Circulation. 2018;138:2227–2242. doi: 10.1161/CIRCULATIONAHA.118.035202
43.
Nomura S, Satoh M, Fujita T, Higo T, Sumida T, Ko T, Yamaguchi T, Tobita T, Naito AT, Ito M, et al. Cardiomyocyte gene programs encoding morphological and functional signatures in cardiac hypertrophy and failure. Nat Commun. 2018;9:4435. doi: 10.1038/s41467-018-06639-7
44.
Frey N, Katus HA, Olson EN, Hill JA. Hypertrophy of the heart: a new therapeutic target?. Circulation. 2004;109:1580–1589. doi: 10.1161/01.cir.0000120390.68287.bb
45.
Zhong Z, Liang S, Sanchez-Lopez E, He F, Shalapour S, Lin XJ, Wong J, Ding S, Seki E, Schnabl B, et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature. 2018;560:198–203. doi: 10.1038/s41586-018-0372-z
46.
Schultz HD, Ustinova EE. Capsaicin receptors mediate free radical-induced activation of cardiac afferent endings. Cardiovasc Res. 1998;38:348–355. doi: 10.1016/s0008-6363(98)00031-5
47.
Scanzano A, Cosentino M. Adrenergic regulation of innate immunity: a review. Front Pharmacol. 2015;6:171. doi: 10.3389/fphar.2015.00171
48.
Rapacciuolo A, Esposito G, Caron K, Mao L, Thomas SA, Rockman HA. Important role of endogenous norepinephrine and epinephrine in the development of in vivo pressure-overload cardiac hypertrophy. J Am Coll Cardiol. 2001;38:876–882. doi: 10.1016/s0735-1097(01)01433-4
49.
Triposkiadis F, Karayannis G, Giamouzis G, Skoularigis J, Louridas G, Butler J. The sympathetic nervous system in heart failure physiology, pathophysiology, and clinical implications. J Am Coll Cardiol. 2009;54:1747–1762. doi: 10.1016/j.jacc.2009.05.015
50.
Fukuda K, Kanazawa H, Aizawa Y, Ardell JL, Shivkumar K. Cardiac innervation and sudden cardiac death. Circ Res. 2015;116:2005–2019. doi: 10.1161/CIRCRESAHA.116.304679
Information & Authors
Information
Published In
Copyright
© 2022 American Heart Association, Inc.
Versions
You are viewing the most recent version of this article.
History
Received: 18 May 2022
Accepted: 11 October 2022
Published online: 28 November 2022
Published in print: 24 January 2023
Keywords
Subjects
Authors
Disclosures
Disclosures None.
Sources of Funding
This work was supported by a Japan Heart Foundation/Novartis Grant for Research Award on Molecular and Cellular Cardiology (to Dr Higashikuni), research grants from the Japan Foundation for Applied Enzymology (to Dr Higashikuni), the Cardiovascular Research Fund (to Dr Higashikuni), the Okinaka Memorial Institute for Medical Research (to Dr Higashikuni), the Fugaku Fund for Medical Pharmaceuticals (to Dr Higashikuni), the Takeda Science Foundation (to Drs Higashikuni, Fukuda, and Sata), the SENSHIN Medical Research Foundation (to Dr Fukuda), and the Vehicle Racing Commemorative Foundation (to Dr Sata), and JSPS KAKENHI Grants (Number JP25860586 and JP15K09133 to Dr Higashikuni, Number JP19K08584 to Dr Fukuda, and Number JP19H03654 and JP22H03069 to Dr Sata).
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.
- MLKL triggers NLRP3 activation in sodium arsenite-induced myocardial necroinflammation, Toxicology, 515, (154132), (2025).https://doi.org/10.1016/j.tox.2025.154132
- Current anti-inflammatory strategies for treatment of heart failure: From innate to adaptive immunity, Pharmacological Research, 216, (107761), (2025).https://doi.org/10.1016/j.phrs.2025.107761
- Dysfunctional cardiomyocyte signalling and heart disease, Current Opinion in Cell Biology, 94, (102517), (2025).https://doi.org/10.1016/j.ceb.2025.102517
- Coordination of innate immune responses by connexins, Frontiers in Immunology, 16, (2025).https://doi.org/10.3389/fimmu.2025.1594015
- Checkpoint kinase Wee1 activation drives inflammation and hypertrophy through the protein kinase B/phosphoinositide 3-kinases–nuclear factor κB pathway in cardiomyocytes, European Heart Journal, (2025).https://doi.org/10.1093/eurheartj/ehaf349
- NLRP3 Inflammasome Triggers Inflammation of Obstructive Sleep Apnea, Current Molecular Medicine, 25, 5, (522-536), (2025).https://doi.org/10.2174/0115665240294605240426123650
- Sauchinone preserves cardiac function in doxorubicin-induced cardiomyopathy by inhibiting the NLRP3 inflammasome, Phytomedicine, 140, (156624), (2025).https://doi.org/10.1016/j.phymed.2025.156624
- Ginseng in delaying brain aging: Progress and Perspectives, Phytomedicine, 140, (156587), (2025).https://doi.org/10.1016/j.phymed.2025.156587
- Sympathetic hyperinnervation drives abdominal aortic aneurysm development by promoting vascular smooth muscle cell phenotypic switching, Journal of Advanced Research, 71, (383-398), (2025).https://doi.org/10.1016/j.jare.2024.05.028
- Mitigation of Doxorubicin Cardiotoxicity With Synergistic miRNA Combinations Identified Using Combinatorial Genetics en masse (CombiGEM), JACC: CardioOncology, (2025).https://doi.org/10.1016/j.jaccao.2025.03.007
- See more
Loading...
View Options
Login options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginPurchase Options
Purchase this article to access the full text.
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.