Skip to main content
International Journal of Molecular Medicine logoLink to International Journal of Molecular Medicine
. 2025 Apr 4;55(6):86. doi: 10.3892/ijmm.2025.5527

Research progress in myocardial function and diseases related to muscarinic acetylcholine receptor (Review)

Chuqiao Shen 1, Qiang Zuo 2, Zhengbin Shao 2, Yixuan Lin 3, Shuo Chen 4,
PMCID: PMC12005369  PMID: 40183403

Abstract

Muscarinic acetylcholine (ACh) receptors (also known as M receptors) are widely distributed in all organs and tissues of the body, mainly playing a role in cholinergic nerve conduction. There are five known subtypes of muscarinic ACh receptors, but their pharmacological mechanisms of action on myocardial function have remained to be clearly defined. Functional myocardial diseases and myocardial injuries, such as arrhythmia, myocardial ischemia, myocarditis and myocardial fibrosis, may be affected by muscarinic ACh receptors. This article reviews the research progress of the regulation of myocardial function by muscarinic ACh receptors and related diseases, with the aim of developing better strategies and providing references for further revealing and clarifying the signal transduction and mechanisms of muscarinic ACh receptors in cardiomyocytes, and finding potential myocardial protective drugs that act on muscarinic ACh receptors.

Key words: muscarinic acetylcholine receptor, myocardial function, myocardial disease, myocardial toxicity, non-neuronal cholinergic system

1. Introduction

The muscarinic acetylcholine (ACh) receptor (M receptor or mAChR), also known as the muscarinic ACh receptor, is an important type of neurotransmitter receptor widely expressed in various tissues and organs of the human body. As a G protein-coupled membrane receptor, it regulates different intracellular signaling transduction mechanisms in the body (1), and has become a target for various chemical drugs (2,3). The cholinergic receptors can be divided into muscarinic ACh receptors and nicotinic ACh receptors (N receptor), and the majority of them in the heart are muscarinic ACh receptors. Therefore, studying the function and disease regulation of muscarinic ACh receptors in the heart is particularly important. Currently, there are a total of five subtypes of muscarinic ACh receptors, which are named M1-M5, and only three subtypes of muscarinic ACh receptors with biological functions have been discovered, which are M1, M2 and M3 (4). According to literature reports, these three functional muscarinic ACh receptors play an important role in the physiological and pathological regulation of the heart (5). However, researchers still have not fully clarified the myocardial function and pathophysiological mechanisms mediated by muscarinic ACh receptors. Known regulation of muscarinic ACh receptors in developing cardiomyopathy involves pathological changes such as myocardial infarction, myocardial fibrosis, cardiac hypertrophy, myocardial contraction and arrhythmia, as well as regulation of myocardial inflammation and non-nerve ACh systems (6). These cardiomyopathies are commonly affected by myocardial apoptosis and necrosis, oxidative stress, mitochondrial dysfunction and autophagy, fission and biogenesis, etc. The exact mechanism by which muscarinic receptors affect these cardiomyopathies is still worthy of further in-depth research. For the present review, studies on recent research progress related to the impact of muscarinic ACh receptors on myocardial function were collected, aiming to discover the potential signaling transduction mechanism of muscarinic ACh receptors in the cardiomyocytes and providing a theoretical basis for further studying myocardial protective drugs that act on muscarinic ACh receptors.

2. The regulatory effects of muscarinic ACh receptors on myocardial contraction

The muscarinic ACh receptor is an important signal transduction mediator present on the membrane of cardiomyocytes, which regulates myocardial contraction (7-9). Over the years, the regulatory mechanism of the M2 muscarinic ACh receptor in cardiomyocytes has been widely reported. It has been pointed out that ACh, phthalimide-azo-iperoxo and naphthalimide-azo-iperoxo can reduce the myocardial contraction force and cardiac output by binding to the M2 muscarinic ACh receptor of cardiomyocytes (10,11). A study also found that bile acids have a new function of slowing down myocardial contraction through the M2 muscarinic ACh receptor (12). Another study reported that an agonist or inhibitor of the M2 muscarinic ACh receptor can regulate the contraction function of the heart by regulating Ras homolog family member A and Rac family small GTPase 1 through the long isoform of the regulator of G protein signaling 3 (13). On the contrary, M2 muscarinic ACh receptor inhibitors such as scopolamine, N-methylscopolamine and [(3R,4R)-3-(3cyclopropyl-1,2,4,oxadiazol-5-yl)-1-azabicyclo[2.2.1]heptane] (L-687,306), have been studied and shown to have the effect of increasing the heart rate and myocardial contraction in rats (14). The calcium channel in cardiomyocytes has long been considered an important pathway for regulating myocardial contraction. It has been reported that ACh can regulate calcium ions in cardiomyocytes, affecting the actomyosin interaction mechanism and the stretching of the sarcomere, thereby changing the myocardial contraction function (15). Furthermore, myocardial diseases are highly associated with the functional obstacle of ryanodine receptor (RyR2). It has been reported that the muscarinic ACh receptor can reduce the stimulation of the parasympathetic nervous system and calcium ion/calcium-dependent protein kinase II (CaMKII)-dependent reactivity, decrease the phosphorylation of RyR2 Ser-2814, lead to increased systolic calcium ion release and reduce the leakage of abnormal calcium ions, thereby improving calcium ion cycling efficiency (16). In addition, it has been proven that muscarinic ACh receptors can regulate the L-type calcium signal channel, CaMKII and the phosphoinositide 3-kinase (PI3K)-protein kinase B (AKT)-neuronal nitric-oxide synthase signaling pathway involved in cardiomyocyte contraction function (17,18). In all, the muscarinic ACh receptor controls the homeostasis of calcium ion currents in cardiomyocytes. The energy metabolism of the heart is also one of the important functions affected by muscarinic ACh receptors. The response to M2 receptor activation could be observed when the agonist stimulates cyclic adenosine monophosphate (cAMP) production (19). There are relevant research reports that the non-selective muscarinic ACh receptor inhibitor atropine can enhance myocardial contraction by blocking cAMP-specific phosphodiesterase type 4 (20). The changes in cAMP affect protein kinase A-dependent phosphorylation targets, such as L-type calcium channels, which play an important role in regulating myocardial contractility (19). Ursolic acid could enhance muscarinic ACh receptor-mediated atrial natriuretic peptide secretion and regulate myocardial contraction (21). It has been reported that ACh can regulate the contractility of atrial myocardium by activating muscarinic receptors and regulating the kinetics of the actin myosin interaction in cardiomyocytes (15). The release of ACh between cardiomyocytes can be affected by neural and drug regulation (22,23). Muscarinic ACh receptors are also involved in the activation of atrial cyclo-oxygenase-2 and autoantibody mediates the positive/negative inotropic response to muscarinic agonists (24,25). In the heart muscle, the function of the muscarinic ACh receptor is closely related to the beating and stopping of the heart. It has been suggested that stimulating the muscarinic ACh receptor can lead to sudden cardiac arrest, while muscarinic ACh receptor inhibitors can effectively protect against this symptom (26). Age can regulate the activity of muscarinic ACh receptors on cardiomyocytes and ultimately affect myocardial contractility. As age increases, the activity of the muscarinic ACh receptor decreases, ultimately affecting myocardial contraction (27-29). The aforementioned research indicates that the muscarinic ACh receptor can regulate myocardial contraction of the heart, laying a foundation for the study of pathological effects of the muscarinic ACh receptor. The effects of muscarinic ACh receptors on myocardial contraction are listed in Table I and shown in Fig. 1.

Table I.

Effects of muscarinic ACh receptors on myocardial contraction.

Muscarinic receptor type Target Agonist/antagonist CVDs Publication year (Refs.)
M2 Ca2+ channel Ang-(1-7), ACh Arrhythmia 2022 (9)
Ca2+ channel PAI Arrhythmia 2019 (10)
cAmp, IK Atropine Dilated cardiomyopathy, ischemic cardiomyopathy, atrial tachyarrhythmias 2011 (25)
Sympathetic nerve Yridostigmine, donepezil Heart failure, hypertension 2024 (23)
RhoA, Racl, RGS3L Carbachol Heart failure 2022 (13)
Parasympathetic nerve Scopolamine, NMS, L-687306, arecoline Bradycardia 2020 (14)
Ca2+ ion channel ACh Atrial fibrillation 2022 (15)
RyR2, PI3K Carbachol Heart failure 2020 (16)
cAMP Bile acid Arrhythmias 2018 (12)
cAMP Carbachol, ACh Cardiac arrest 2018 (26)
CSPG4 Carbachol Arrhythmia 2024 (27)
M3 L-type calcium channel, CaMKII Tiotropium bromide Myocardial infarction 2019 (18)
Caspase-1, IL-1β 4-DAMP Myocardial infarction, heart failure 2018 (29)
M1, M2 Parasympathetic nerve Pirenzepine, atropine Bradycardia 1997 (28)
M2, M3 RyR2 AF-DX116, J104129 Heart failure 2016 (17)
Cyclo-oxygenase-2 Carbachol Heart failure 2014 (24)
M1, M2, M3 PKA-dependent phosphorylation, L-type calcium channel cAMP Ischemia-reperfusion injury 2024 (19)
Vagal afferent nerve ACh Heart failure 2020 (22)
PDE4 Atropine Tachycardia, arrhythmias 2017 (20)
ANP secretion Ursolic acid Myocardial infarction 2014 (21)

ACh, acetylcholine; M, muscarinic ACh receptor; ACh, acetylcholine; cAMP, cyclic adenosine monophosphate; RGS3L, long isoform of the regulator of G protein signaling 3; CSPG4, chondroitin sulfate proteoglycan 4; RyR2, ryanodine receptor; PI3K, phosphoinositide 3-kinase; CaMKII, calcium ion/calcium-dependent protein kinase II; IL, interleukin; PDE4, phosphodiesterase 4; PKA, protein kinase A; RhoA, Ras homolog family member A; Rac1, Rac family small GTPase 1; ANP, atrial natriuretic peptide; PAI, phthalimide-azo-iperoxo; Ang-(1-7), angiotensin-(1-7); NMS, N-methylscopolamine; 4-DAMP, 4-diphenylacetoxy-N-methylpiperidine methiodide; L-687306, (3R,4R)-3-(3-cyclopropyl-1,2,4-oxadiazol-5-yl)-1-azabicyclo[2.2.1]heptane; AF-DX116, 11-[[2-[(diethylamino)methyl]-1-piperidinyl]acetyl]-5,1 1-dihydro-6H-pyrido[2,3-b][1,4] benzodiazepin-6-one; J104129, (αR)-α-cyclopentyl-α-hydroxy-N-[1-(4-methyl-3-penten-1-yl)-4-piperidinyl]-benzeneacetamide fumarate.

Figure 1.

Figure 1

Regulatory effect of the muscarinic ACh receptor on myocardial contraction. The numerous cellular mechanisms include the activation of calcium and potassium channels, nerve and signal proteins, changes in biochemical indicators and changes in inflammatory factors, which may be involved in the contractile function of myocardium. M, muscarinic ACh receptor; ACh, acetylcholine; cAMP, cyclic adenosine monophosphate; RGS3L, long isoform of the regulator of G protein signaling 3; CSPG4, chondroitin sulfate proteoglycan 4; RyR2, ryanodine receptor; PI3K, phosphoinositide 3-kinase; CaMKII, calcium ion/calcium-dependent protein kinase II; IL, interleukin; PDE4, phosphodiesterase 4; PKA, protein kinase A.

3. The regulatory effects of muscarinic ACh receptors on myocardial infarction

The impact of the muscarinic ACh receptor on myocardial infarction is significant. Acute myocardial infarction is the risk disease with the highest mortality rate among patients with heart disease. The underlying pathogenesis and influencing factors have been the focus of medical research for numerous years (30). In myocardial infarction, the muscarinic ACh receptor can affect the onset and course of myocardial infarction. Meanwhile, the muscarinic ACh receptor in the cardiomyocytes can express increased expression in the conduction block caused by myocardial infarction (31). Some studies have pointed out that muscarinic ACh receptor inhibitors can worsen the condition of myocardial infarction by inhibiting the muscarinic ACh receptor on the cardiomyocyte (32). In basic and clinical research on myocardial infarction, it has been shown that the regulation of muscarinic ACh receptors can induce the occurrence of myocardial infarction, which in turn leads to heart failure (18,33). The mechanism of action of muscarinic ACh receptors mediating myocardial infarction is relatively complex, and it may involve multiple signaling pathways (34). Among them, the regulation of calcium channels is the main reason for triggering myocardial contraction and affecting the disease process of myocardial infarction. It has been shown that the receptor-interacting protein 3-induced activation of CaMKII in cardiomyocytes can trigger oxidative stress in cardiomyocytes, which has always been an important cause of myocardial infarction (35). This may be caused by affecting the opening of the mitochondrial permeability transition pore and myocardial necrosis. In response to the above mechanism of myocardial infarction injury, the pharmacological community has carried out corresponding research on the protective effects of muscarinic ACh receptors. Certain studies have reported that the agonist effect of drugs on muscarinic ACh receptors can alleviate myocardial infarction (36,37). Agitating muscarinic ACh receptors can protect against myocardial infarction through extracellular signal-regulated kinase 1/2 (ERK1/2)- and PI3K/AKT-mediated signaling pathways (38), and play a protective role in hypoxia and injury of cardiomyocytes (39). ACh can also alleviate the injury of cardiomyopathy by affecting adenosine 5′-monophosphate-activated protein kinase (AMPK) signaling and mitochondrial cristae reconstruction through interaction with muscarinic ACh receptors (40). Studies have shown that ACh can reduce the transient calcium amplitude of muscarinic ACh receptors to regulate the levels of calcium and iron, thereby alleviating oxidative stress injury (41). Vagal nerve stimulation experiments have shown that the increase in ACh levels can upregulate the expression of vascular endothelial growth factor A/B, and promote angiogenesis and protection, thereby playing a protective role in myocardial infarction (42,43). This result was later confirmed by the repair of the injured coronary artery and cardiomyocytes during infarction through the M2 muscarinic ACh receptor (44). Studies have shown that acetylcholinesterase (AChE) inhibitors can better protect against myocardial ischemia, myocardial infarction and heart failure than simply stimulating the release of ACh (45,46).

In recent years, the M3 muscarinic ACh receptor, as an important regulatory receptor in cardiomyocytes, has received widespread attention from the cardiovascular research community. Another study showed that inhibiting microRNA (miR)-376b-5p can affect the activation of the M3 muscarinic ACh receptor, thereby regulating downstream calcium signaling and reactive oxygen species-related cardioprotection pathways (47). Zhao et al (48) reported that agonist activation of the M3 muscarinic ACh receptor can alleviate myocardial injury caused by myocardial ischemia by regulating connexin 43 (Cx43) phosphorylation and cyclooxygenase-2 expression. Another study showed that chlorine-based choline can alleviate myocardial injury under the transverse aortic constriction myocardial ischemic model by agonist activation of the M3 muscarinic ACh receptor, while the M3 muscarinic ACh receptor inhibitor 4-diphenylacetoxy-N-methylpiperidine methiodide had the opposite effect (49,50). Inhibiting the M3 muscarinic ACh receptor was also observed to promote the influx of calcium ions into cells after myocardial ischemia, interfering with normal cellular energy metabolism and accelerating the death of cardiomyocytes (18). Wang et al (51) reported the signaling role of beta-catenin in the regulation of the downstream anti-apoptotic protein Bcl-2 mediated by the M3 muscarinic ACh receptor. Therefore, the regulatory role of the M3 muscarinic ACh receptor on myocardial infarction is relatively certain (Fig. 2).

Figure 2.

Figure 2

Regulatory mechanisms of muscarinic ACh receptor on myocardial infarction. ACh and miR-376b-5p can act on muscarinic receptors, affect HIF-1α through MAPK/ERK1/2, Cx43/Cox2 and PI3K/Akt, and affect cardiomyocyte apoptosis, autophagy, endoplasmic reticulum stress and mitochondrial biogenesis, and ultimately affect myocardial infarction. ACh, acetylcholine; M, muscarinic acetylcholine receptor; mAChR, muscarinic ACh receptor; MAPK, phospho-p38 mitogen-activated protein kinase; ERK1/2, extracellular signal-regulated kinase 1/2; Cx43, connexin 43; Cox-2, cyclooxygenase-2; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; HIF-1α, hypoxia inducible factor-1α; ER, endoplasmic reticulum; miR, microRNA; p-AKT, phosphorylated AKT.

4. Effects of muscarinic ACh receptors on myocardial ischemia and reperfusion injury

Existing studies have shown that ischemia/reperfusion injury is closely related to the regulation of muscarinic ACh receptors (Fig. 3). According to relevant research reports, various anti-muscarinic ACh receptor drugs, including ipratropium, tiotropium bromide, atropine and adriamycin, have been reported to increase the incidence of ischemic/reperfusion myocardial injury by regulating the muscarinic ACh receptor, resulting in cardiac toxicity (18,52,53). ACh can play a protective role in ischemic reperfusion injury by stimulating the muscarinic ACh receptor. Therefore, exploring the protective mechanism of the muscarinic ACh receptor against ischemic reperfusion injury is becoming a new research hotspot in the field of myocardial ischemia/reperfusion injury. There is evidence that by stimulating the muscarinic ACh receptor on the heart, it can have a significant protective effect on the mouse myocardial ischemia-reperfusion injury model, reducing the area of myocardial injury caused by ischemia-reperfusion (34). It was also shown that stimulation of the myocardial muscarinic ACh receptor can cause the release of ACh mediated by the muscarinic ACh receptor, resulting in a response of ischemic preconditioning, which can protect against myocardial ischemia-reperfusion injury (54). Donepezil and other cholinergic receptor agonists can protect against cell apoptosis after myocardial ischemia-reperfusion injury by regulating the level of phosphorylated (p)-Cx43 (ser368) and the balance of mitochondrial activity and autophagy (55). ACh can also alleviate endoplasmic reticulum stress in myocardial cells and increase cell viability (56), blocking mitochondrial unfolding protein, thereby reducing myocardial cell apoptosis induced by hypoxia/reoxygenation (57). Existing studies have shown that vagus nerve stimulation activating the muscarinic ACh receptor can reduce mitochondrial function through the M3 muscarinic ACh receptor/CaMK kinase b/AMPK signaling pathway to protect against myocardial ischemic injury (58). The mechanism of this ischemic myocardial injury may be myocardial cell apoptosis and related metabolic dysfunction. Experiments with catestatin binding to the M2 muscarinic ACh receptor indicate that the M2 muscarinic ACh receptor can play a myocardial protective role by regulating the ERK1/2 and PI3K/AKT signaling pathways in cells (38).

Figure 3.

Figure 3

Role of muscarinic ACh receptor in myocardial ischemia and reperfusion injury. Ischemia/reperfusion injury may be regulated by drugs and stimuli through m2AChR receptors on PI3K/Akt and ERK1/2 pathways. Meanwhile, ischemia/reperfusion injury may be affected by ACh and drugs through m3AChR on cardiomyocyte apoptosis, autophagy, ER stress and mitochondrial biogenesis regulated by the AMPK pathway. ACh, acetylcholine; mAChR, muscarinic ACh receptor; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; p-AKT, phosphorylated AKT; ERK1/2, extracellular signal-regulated kinase 1/2; CaMKKb, calmodulin-dependent protein kinase kinase b; ER, endoplasmic reticulum.

5. Effects of muscarinic ACh receptors on myocardial fibrosis

Myocardial fibrosis can occur in various cardiovascular diseases and has been considered one of the most common conditions in cardiomyopathy, the pathogenesis of which remains to be fully elucidated. Although cardiac fibroblasts are the main cells that constitute heart tissue, the mechanism of muscarinic ACh receptor-induced myocardial fibrosis still requires further research (Fig. 4). In addition, relevant neural studies have shown that stimulation of the vagus nerve, which is the same as the parasympathetic nerve, and exogenous stimulation of the M3 muscarinic ACh receptor, can alleviate the process of myocardial fibrosis via the miR-29b/beta-site app cleaving enzyme 1 axis both in vivo and in in vitro experimental models (49,59). An in vitro experimental study showed that inhibiting ACh levels can induce the formation of cardiac fibroblasts (60). A previous clinical study has shown that the amount of M2 muscarinic ACh receptor autoantibody can be used as a suitable detection method for judging the severity of the pathological and physiological conditions of patients with left atrial fibrosis (61). In addition, recent experimental results of a human mechanistic study also showed that the level of M2 muscarinic ACh receptor autoantibody in the serum of patients with atrial fibrosis was significantly higher than that in the non-atrial fibrosis group, and immunohistochemical analysis and western blot analysis of left atrial appendage tissue suggested that M2 muscarinic ACh receptor is closely related to the process of myocardial fibrosis (62). All of the above evidence indicated that the muscarinic ACh receptor has an important connection with the onset, diagnosis and treatment of myocardial fibrosis. Another study showed that the M1 muscarinic ACh receptor is significantly upregulated in patients with chronic atrial fibrillation and can affect atrial fibrillation by regulating the IK,Ach channel in human atrial myocytes (63).

Figure 4.

Figure 4

Mechanism of muscarinic ACh receptor in myocardial fibrosis. Stimulus and autoantibodies may affect myocardial fibrosis through ACh and potassium channels of M1, M2 and M3 receptor-mediated microRNA-29b/BACE1. M, muscarinic ACh receptor; ACh, acetylcholine; BACE1, beta-site app cleaving enzyme 1.

6. Effects of muscarinic ACh receptors on arrhythmia

Recently, it has been shown that arrhythmia is related to the regulation of muscarinic ACh receptors (Fig. 5). Activation of the muscarinic ACh receptor can reduce heart rate and even cause bradycardia (64). Studies have confirmed that blocking the parasympathetic nerve in the heart is an important factor in inducing ventricular arrhythmia and increasing myocardial energy consumption (65). In the study of arrhythmia, the regulatory mechanism of IK channels has recently attracted attention. Studies have shown that agonists of muscarinic ACh receptors, including ACh, can alleviate the symptoms of cardiac arrhythmia without affecting the contractility of the heart (66). The mechanism includes hypoxia and regulation of IK-ATP (67-69). Atrial fibrillation is one of the important symptoms in patients with arrhythmia, and it is closely related to the regulatory effect of muscarinic ACh receptors on heart function. Therefore, numerous scholars have conducted in-depth research on the association between atrial fibrillation and myocardial muscarinic ACh receptors. It has been shown that inhibiting the release of ACh can effectively inhibit the effect of atrial fibrillation in patients after cardiac surgery (70). The specific manifestation showed that M2 muscarinic Ach receptors could induce the release of autoantibodies and thyroid hormones, promoting the susceptibility to atrial fibrillation and sinus tachycardia (27,71). Researchers have found that the level of autoantibodies to M2 muscarinic ACh receptors in patients with atrial fibrillation shows a significant increase (62). Another study reported that changes in the voltage sensitivity of M2 muscarinic ACh receptors can lead to heart diseases such as atrial fibrillation and sinus tachycardia (72). Furthermore, M3 muscarinic ACh receptor overexpression was observed to reduce the incidence and mortality of arrhythmia after myocardial ischemia-reperfusion injury. This effect may be mediated by downregulating the expression of arrhythmogenic miR-1 and increasing the inward rectifier potassium current, which may be a new anti-arrhythmic strategy for diagnosis (73). Other studies have shown that the release of ACh triggered by parasympathetic nerve cells can increase the sensitivity of animals to arrhythmia and increase the heart rate (74-76).

Figure 5.

Figure 5

Effect of muscarinic ACh receptor on arrhythmia. ACh may protect against atrial fibrillation and sinus tachycardia caused by the increase of autoantibodies and thyroid hormones through M2 receptors induced by voltage. Both M2 and M3 could regulate cardiomyocyte ATP, miR-1 and hypoxia through potassium ion channels to affect arrhythmia. M, muscarinic ACh receptor; ACh, acetylcholine; miR, microRNA.

7. Effects of muscarinic ACh receptors on heart failure and myocardial hypertrophy

An increasing number of studies have shown that stimulating the muscarinic ACh receptor through drugs or neural transmission may become an important means of treating heart failure (77) (Fig. 6). Certain studies have shown that the continuous excitation of muscarinic ACh receptor by ACh can release calcium ions through the RyR2-mediated calcium store of the sarcoplasmic reticulum in cardiomyocytes, and the regulation of calcium signals related to store-operated calcium entry can alleviate heart failure (78,79). Another study has shown that ACh can serve as a key compensatory mediator in the development of heart failure in mice by stimulating the M2 muscarinic ACh receptor of cardiomyocytes to alleviate the occurrence and development of ventricular remodeling and heart failure (80,81). The drug donepezil, which has the function of AChE inhibitor, can increase ACh in rat hearts and play a long-term protective effect on heart failure (82). Research data show that increasing ACh by using central or peripheral AChE inhibitors can effectively improve heart failure and the heart's autonomic nervous imbalance and hemodynamic changes in patients with hypertension (83). ACh can regulate the effect of AMPK on mitochondrial cristae remodeling by stimulating the muscarinic ACh receptor, thereby alleviating the hypertrophy of cardiomyocytes induced by palmitic acid (40).

Figure 6.

Figure 6

Effect of muscarinic ACh receptor on heart failure and myocardial hypertrophy. (A) Effect of muscarinic ACh receptor on heart failure. (B) Effect of muscarinic ACh receptor on myocardial hypertrophy. M, muscarinic ACh receptor; ACh, acetylcholine; AMPK, adenosine 5′-monophosphate-activated protein kinase; SOCE, store-operated calcium entry; RyR2, ryanodine receptor; MAPK, mitogen-activated protein kinase.

Myocardial hypertrophy is a common clinical cardiomyopathy, which is also closely related to the induction of muscarinic ACh receptor (84). One of the reasons for myocardial hypertrophy may be related to the decrease in K+ repolarization associated with the M3 muscarinic ACh receptor, which leads to the harmful myocardial remodeling. Overexpression of M3 muscarinic ACh receptor in cardiomyocytes can alleviate the adverse myocardial remodeling (85). Wang et al (86) found that cardiac hypertrophy can play a protective role by inhibiting the P38/p-p38 mitogen-activated protein kinase signaling pathway and blocking the M3 muscarinic ACh receptor. The role of M2 muscarinic ACh receptor in regulating myocardial hypertrophy is also relatively obvious. Studies show that aerobic exercise can improve myocardial hypertrophy by regulating M2 muscarinic ACh receptors, affecting mitochondrial quality control and endoplasmic reticulum stress in cardiomyocytes (87). It has been shown that autoantibodies to M2 muscarinic ACh receptor can also cause myocardial hypertrophy in rabbits (88). The AChE inhibitor pyridostigmine can improve myocardial hypertrophy by prolonging the duration of ACh on the M2 muscarinic ACh receptor. These results were confirmed by echocardiography, immunofluorescence and precipitation methods (89). Intermittent excessive activation of M2 muscarinic ACh receptors can also exacerbate myocardial hypertrophy through oxidative stress (90).

8. Effects of muscarinic ACh receptors on myocardial inflammation

The regulation of the inflammatory response by the muscarinic ACh receptor has also been widely studied (Fig. 7). The role of the muscarinic ACh receptor in regulating myocardial inflammation is mainly achieved by affecting neural transmission and changes in lymphocytes. Studies have shown that, compared to the normal group, myocardial inflammation confirmed by enzyme-linked immunosorbent assay is closely related to the increase in the level of self-antibodies mediated by the M2 muscarinic ACh receptor (91), and the autoimmune antibodies show a role in heart function damage and myocardial inflammation (92). Through stimulating cholinergic neurons, a study showed good anti-inflammatory effects in a heart model of Wistar rats through immunohistochemistry and cytokine measurement (93). In addition, a study suggested that the inhibition of AChE by bromocriptine enhances the function of ACh, thereby preventing inflammatory autonomic dysfunction (94). The muscarinic ACh receptor can also regulate the inflammatory immune mediators of myocardial cells, such as the anti-inflammatory effect of T lymphocytes (95). Cox et al (96) reported that infection can promote the increase in the expression of ACh transferase (ChAT) in CD4+T and CD8+T cells, thereby enhancing the immune response. Furthermore, the release of ACh and ChAT+ B lymphocytes has been shown to inhibit the activation of macrophages (97). In the viral myocarditis mouse model, the anti-inflammatory pathway of ACh may be achieved through the differentiation of CD4+ T cells and the regulation of the expression of Th1 and Th17 cytokines (98). This may reveal the current regulatory mechanism of myocardial inflammation, such as myocardial cell lesions and cell infiltration symptoms. Exogenous ACh stimulation may activate the PI3K/AKT pathway (99), and the ERK/early growth response-1 pathway (100,101), thereby alleviating inflammation and oxidation. Similarly, it has been found that a small population of ChAT+ natural killer cells, which are, however, transcriptionally distinct, have immune protective effects in a mouse model (102). The stimulation of the muscarinic ACh receptor can also alleviate cardiac inflammation through the muscarinic ACh and nicotine receptors. Recently, the regulatory effect of the non-neuronal cholinergic system on myocardial inflammation has also been reported (97). A study suggested that the non-neuronal cholinergic system can regulate the activation and inhibition of inflammatory cells, such as the number of macrophages and Forkhead box protein P3+ T cells in myocardium through the muscarinic ACh receptor (103). However, studies have indicated that, although the non-neuronal cholinergic system can regulate the release of ACh and produce the effect of stimulating the muscarinic ACh receptor, it was not found to improve or exacerbate the myocardial inflammatory response (104,105). Therefore, whether the non-neuronal cholinergic system can play a role in the myocardial inflammatory response still requires further research. All of the roles of myocardial inflammation through the regulation of muscarinic ACh receptors are listed in Table II.

Figure 7.

Figure 7

Role of muscarinic ACh receptor on myocardial inflammation. Drugs, chemicals and biological mediators may act on muscarinic ACh receptors to regulate various immune responses, affecting viral myocarditis, cardiac output and endothelial adhesion. The inflammatory responses could affect the regulation of myocardial injury through the ERK, PI3K/AKT, oxidative stress, mitochondrial function and angiogenesis pathways on the regulation of myocardial injury. M, muscarinic ACh receptor; ACh, acetylcholine; LPS, lipopolysaccharide; IL, interleukin; TNF, tumor necrosis factor; Egr-1, early growth response-1; ERK, extracellular signal-regulated kinase; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; CD, cluster of differentiation; FOXP3, forkhead box p3; SDF-1α, stromal cell-derived factor-lα; IFN-γ, interferon-γ; ChAT, acetylcholine transferase; Th, helper T cell; DM2R-el2, donor-immunized second extracellular loop of M2R; RM2R-il3, recipient third intracellular loop of M2R; MYH7, myosin heavy chain 7; TNNT2, troponin T2; CVB3, Coxsackievirus B3.

Table II.

Mechanisms of muscarinic ACh receptors in myocardial inflammation.

Function Stimulus Species Target Mechanism Year (Refs.)
Hypertrophic cardiomyopathy M2 autoantibody Homo sapiens C-reactive protein, high-sensitivity C-reactive protein MYH7 and TNNT2 genetic heterogeneity 2020 (91)
RM2R-il3, DM2R-el2 Mouse Self-antibodies Autophagy and mitophagy 2018 (92)
ACh, LPS Mouse Vesicular ACh transporter Monocytes/macrophages, T cells, B cells, neutrophils 2021 (103)
Pyridostigmine, Ach Mouse TNF-α, IL-6, IL-1β, ERK, Egr-1 Metabolic inflammation, cardiac lipotoxicity 2018, 2023 (100,101)
MI Pyridostigmine bromide Rat CD68, CD206, FOXP3, IL-1β, IL-6, IL-10, TNF-α Parasympathetic neuro-immune, oxidative stress 2017 (93)
Pyridostigmine bromide Rat IFN-γ, IL-6, IL-1β, IL-10, TNF-α Mitochondrial dysfunction, inflammatory cell influx, angiogenesis 2019 (94)
ACh, mecamylamine, atropine Rat TNF-α, IL-6, IL-1β, CD31, SDF-1α PI3K/AKT 2023 (99)
Viral myocarditis ACh Mouse ChAT, CD4+, CD8+ T cells T-cell migration 2019 (96)
CVB3 Mouse CD4+ T cells, Th1, Th17 cells CD4+ T-cell differentiation 2018 (98)
Endothelial cell adhesion ACh Mouse CD4+ T-cells, ChAT+ B cells Recruitment of neutrophils 2013 (97)
Cardiac output LPS Rat IL-1β, IL-6, TNF-α Cholinergic anti-inflammatory pathway 2018 (104)
ACh Homo sapiens ChAT, CD4+ T cells Vascular endothelial function 2023 (105)

ChAT, choline acetyltransferase; ACh, acetylcholine; LPS, lipopolysaccharide; M, muscarinic ACh receptor; MI, myocardial infarction; ACh, acetylcholine; LPS, lipopolysaccharide; IL, interleukin; TNF, tumor necrosis factor; Egr-1, early growth response-1; ERK, extracellular signal-regulated kinase; PI3K, phosphoinositide 3-kinase; AKT, protein kinase B; CD, cluster of differentiation; FOXP3, forkhead box p3; SDF-1α, stromal cell-derived factor-lα; IFN-γ, interferon-γ; ChAT, acetylcholine transferase; Th, helper T cell; DM2R-el2, donor-immunized second extracellular loop of M2R; RM2R-il3, recipient third intracellular loop of M2R; MYH7, myosin heavy chain 7; TNNT2, troponin T2; CVB3, coxsackievirus B3.

9. Study on the muscarinic ACh receptors and cardiotoxic effects

Long-term clinical studies have found that numerous anti-muscarinic ACh receptor drugs have cardiotoxic effects on the heart (32,106). Long-acting muscarinic ACh inhibitors such as tiotropium and other tracheal dilating drugs have been reported to exacerbate myocardial injury (107,108) and increase the incidence rate of acute coronary syndrome (109). The use of muscarinic ACh receptor inhibitors such as oxybutynin, solifenacin and tolterodine, which are used to treat hair loss, has been found to be associated with the incidence rate of myocardial disease (110). Adriamycin has long been considered to have cardiotoxic effects, but its mechanism of toxicity in the body has remained elusive. In recent years, studies on its regulation of the muscarinic ACh receptor in cardiomyocytes have been ongoing. Research suggests that the activation of the muscarinic ACh receptor can protect against myocardial injury and cardiomyocyte apoptosis caused by adriamycin, mediated by the nuclear factor erythroid 2-related factor 2/heme-oxygenase-1 pathway (111). It has been shown that donepezil can protect against oxidative stress and mitochondrial function deficiency, reduce the apoptotic ratio of Bax/Bcl-2 and cleaved-caspase 3/caspase 3, and protect against receptor-interacting protein kinase 1-mediated necrosis of cardiomyocytes caused by adriamycin by activating the muscarinic ACh receptor (112). Another study indicated that saffron can exert antioxidant properties and inhibit the apoptosis pathway of myocardial cells through the M2 muscarinic ACh receptors, thereby preventing myocardial toxicity caused by organophosphate poisoning (113). Furthermore, increased ACh under parasympathetic stimulation can inhibit the programmed death of cardiomyocytes, protecting against adriamycin-induced myocardial toxicity (114). In addition, high concentrations of bile acids were shown to produce cardiotoxic effects by acting on the M2 muscarinic ACh receptor (12). ACh can promote lipolysis and activate mitochondrial interactions through perilipin 5, effectively inhibiting cardiomyocyte apoptosis and lipotoxicity (115). The release of large amounts of carbachol stimulates ACh to activate the muscarinic ACh receptor, leading to coronary artery narrowing and changes in the structure of left ventricular cardiomyocytes in rats, causing myocardial toxicity, while long-term treatment with cannabinol can significantly inhibit this toxic effect (116). A schematic illustrating the mechanisms of drugs acting on muscarinic ACh receptors with cardiotoxic effects is provided in Fig. 8.

Figure 8.

Figure 8

Muscarinic ACh receptors and regulation of cardiotoxicity. Numerous drugs and chemicals may act on muscarinic ACh receptors and affect the lipid toxicity and oxidative stress of cardiomyocytes through PLIN5, Nrf2 and HO-1. Subsequently, it affects cell apoptosis, necrosis and lipolysis by changing mitochondrial function and RIP1. These mechanisms ultimately affect the state of the coronary artery, myocardial cells and myocardial injury. ACh, acetylcholine; PLIN5, perilipin 5; Nrf2, nuclear factor erythroid 2-related factor 2; HO-1, heme-oxygenase-1; RIP1, receptor-interacting protein kinase 1.

10. Effects of muscarinic ACh receptors on the non-neuronal cholinergic system (NNCS)

With the continuous deepening of the research on muscarinic ACh receptors, a unique cholinergic regulatory factor production and circulatory system was discovered in recent years, which has attracted widespread attention from pharmacologists. The degradation of ACh in the body by AChE also maintains it at an appropriate level, which may be due to the regulatory function of the non-neural cholinergic system (NNCS) (117). In the heart, this is a self-regulating cholinergic synthesis and secretion system of cardiomyocytes, and except for nerve cells, almost all components of the neural ACh system are retained. The NNCS may contain components such as ChAT, ACh, nicotinic/muscarinic ACh receptor, high-affinity choline uptake and cholinesterase. They can participate in cell proliferation, migration, differentiation, cell barrier formation, programmed cell death, hypoxia/reoxygenation injury and other cellular functions and metabolic processes (118-120). The acute promyelocytic leukemia cell line NB-4 can regulate the expression of M3 muscarinic ACh receptors under the stimulation of choline, thus exerting its anti-inflammatory effects in vivo (121). ChAT-transgenic cells have been confirmed to have the potential of systemic anti-inflammatory response, which can alleviate inflammatory lesions (122,123). Cardiomyocytes increase the expression of AChE through the activation of muscarinic ACh receptors and ChAT gene transcription, and then synthesize ACh. Both RNA interference experiments of ChAT gene and M-receptor inhibitor competition experiments have proved this (124). Further research showed that, through the regulation of the NNCS in cardiomyocytes, the expression of ChAT is higher in female hearts than in males. The circadian rhythm has an important impact on histone acetyltransferase activity, thereby regulating the expression of ChAT (125). The newly discovered S-nitroso-N-acetyl-DL-penicillamine can produce nitric oxide and cyclic guanosine monophosphate, activate the expression of the AChE gene and the function of muscarinic ACh receptors, and have a protective effect against myocardial injury (126). The muscarinic ACh receptors on the NNCS are presented in Fig. 9. Although numerous relevant research achievements have been made in this direction in recent years, there are still many gaps and deficiencies in the study of NNCS in cardiomyocytes that need to be filled. In the future, this may provide a new diagnostic and therapeutic method for myocardial infarction and inflammatory protection.

Figure 9.

Figure 9

Mechanisms of muscarinic ACh receptors in the non-neuronal cholinergic system of cardiomyocytes. The cardioprotective mechanism of non-neuronal cholinergic system of cardiomyocytes is based on increasing the production of ACh. This includes stimulation of cholinergic neurons to electrically activate cholinergic neurons to produce and release ACh. Another possibility may be the pharmacological method of prolonging the effect of ACh by using AChE inhibitors. SNAP could activate muscarinic ACh receptor by transmitting nitric oxide and cGMP to cardiomyocytes, thereby increasing the synthesis and expression of ChAT and ACh. Finally, the circadian rhythm of the heart affects the synthesis and expression of ChAT and ACh by regulating HAT. mAChR, muscarinic ACh receptor; ACh, acetylcholine; AChE, acetylcholinesterase; SNAP, S-nitroso-N-acetyl-DL-penicillamine; cGMP, cyclic guanosine monophosphate; HAT, histone acetyltransferase; ChAT, ACh transferase.

11. Conclusion

The muscarinic ACh receptor and cardiac myocytes are both involved in the regulation of heart function. Current research on the myocardial function and diseases related to the muscarinic ACh receptor shows that its range of action is closely related to numerous heart diseases and physiological functions, but its potential mechanism of action is still being continuously explored. So far, the function and structure of the muscarinic ACh receptor have been successively excavated by the research community, but the regulatory role it plays in the function of cardiomyocytes, as well as its physiological and pathological mechanisms of action, still remain to be fully elucidated. This article summarized the research progress on myocardial contraction and related diseases mediated by muscarinic ACh receptors. It was found that muscarinic ACh receptors can play important roles in regulating calcium homeostasis, mitochondrial dysfunction, cardiomyocyte apoptosis and autophagy, inflammatory cells and mediators, providing potential new research directions for the development of cardiomyopathy drugs in the future. With the introduction of the new concept of the myocardial NNCS, the research on the muscarinic ACh receptor in the field of myocardial function and related diseases shows further extensive prospects and value. These findings will provide new theoretical evidence for muscarinic ACh receptors as potential targets for myocardial protective drugs.

Acknowledgements

Not applicable.

Funding Statement

This work was supported by the National Natural Science Foundation of China (grant no. 81904147), the Youth Research Project of Health Commission of Anhui Province (grant no. AHWJ2023A30241), the Scientific Research Foundation of Education Department of Anhui Province of China (grant no. 2024AH051007) and the Scientific Research Foundation of Education Department of Anhui Province of China (grant no. 2023AH050842).

Availability of data and materials

Not applicable.

Authors' contributions

CS was involved in writing the original draft, data curation and conceptualization of the study. QZ contributed to writing the original draft and was responsible for methodology and visualization. ZS and YL were involved in reviewing and editing the manuscript. SC contributed to the review and editing of the manuscript and was responsible for the supervision and conceptualization of the study. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

  • 1.Lymperopoulos A, Cora N, Maning J, Brill AR, Sizova A. Signaling and function of cardiac autonomic nervous system receptors: Insights from the GPCR signalling universe. FEBS J. 2021;288:2645–2659. doi: 10.1111/febs.15771. [DOI] [PubMed] [Google Scholar]
  • 2.Santos R, Ursu O, Gaulton A, Bento AP, Donadi RS, Bologa CG, Karlsson A, Al-Lazikani B, Hersey A, Oprea TI, Overington JP. A comprehensive map of molecular drug targets. Nat Rev Drug Discov. 2017;16:19–34. doi: 10.1038/nrd.2016.230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Maeda S, Qu Q, Robertson MJ, Skiniotis G, Kobilka BK. Structures of the M1 and M2 muscarinic acetylcholine receptor/G-protein complexes. Science. 2019;364:552–557. doi: 10.1126/science.aaw5188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Foster DJ. Muscarinic receptors: From clinic to bench to clinic. Trends Pharmacol Sci. 2022;43:461–463. doi: 10.1016/j.tips.2022.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Saternos HC, Almarghalani DA, Gibson HM, Meqdad MA, Antypas RB, Lingireddy A, AbouAlaiwi WA. Distribution and function of the muscarinic receptor subtypes in the cardiovascular system. Physiol Genomics. 2018;50:1–9. doi: 10.1152/physiolgenomics.00062.2017. [DOI] [PubMed] [Google Scholar]
  • 6.Palma JA. Muscarinic control of cardiovascular function in humans: A review of current clinical evidence. Clin Auton Res. 2024;34:31–44. doi: 10.1007/s10286-024-01016-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Alom F, Miyakawa M, Matsuyama H, Nagano H, Tanahashi Y, Unno T. Possible antagonistic effects of the TRPC4 channel blocker ML204 on M2 and M3 muscarinic receptors in mouse ileal and detrusor smooth muscles and atrial myocardium. J Vet Med Sci. 2018;80:1407–1415. doi: 10.1292/jvms.18-0197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Schoeller C, Hoffmann S, Adolph S, Regenthal R, Abraham G. Expression of muscarinic acetylcholine receptors in turkey cardiac chambers. Res Vet Sci. 2021;136:602–608. doi: 10.1016/j.rvsc.2021.04.016. [DOI] [PubMed] [Google Scholar]
  • 9.Pontes CNR, Scalzo S, Jesus ICG, Jesus EF, Nunes ADC, Mendonça MM, Mendes EP, Colugnati DB, Xavier CH, Pedrino GR, et al. Angiotensin-(1-7) attenuates the negative inotropic response to acetylcholine in the heart. Peptides. 2022;158:170862. doi: 10.1016/j.peptides.2022.170862. [DOI] [PubMed] [Google Scholar]
  • 10.Riefolo F, Matera C, Garrido-Charles A, Gomila AMJ, Sortino R, Agnetta L, Claro E, Masgrau R, Holzgrabe U, Batlle M, et al. Optical control of cardiac function with a photoswitchable muscarinic agonist. J Am Chem Soc. 2019;141:7628–7636. doi: 10.1021/jacs.9b03505. [DOI] [PubMed] [Google Scholar]
  • 11.Woudstra J, Feenstra RGT, Vink CEM, Marques KMJ, Boerhout CKM, de Jong EAM, de Waard GA, van de Hoef TP, Chamuleau SAJ, Eringa EC, et al. Comparison of the diagnostic yield of intracoronary acetylcholine infusion and acetylcholine bolus injection protocols during invasive coronary function testing. Am J Cardiol. 2024;217:49–58. doi: 10.1016/j.amjcard.2024.01.038. [DOI] [PubMed] [Google Scholar]
  • 12.Ibrahim E, Diakonov I, Arunthavarajah D, Swift T, Goodwin M, McIlvride S, Nikolova V, Williamson C, Gorelik J. Bile acids and their respective conjugates elicit different responses in neonatal cardiomyocytes: Role of Gi protein, muscarinic receptors and TGR5. Sci Rep. 2018;8:7110. doi: 10.1038/s41598-018-25569-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Levay MK, Krobert KA, Vogt A, Ahmad A, Jungmann A, Neuber C, Pasch S, Hansen A, Müller OJ, Lutz S, Wieland T. RGS3L allows for an M2 muscarinic receptor-mediated RhoA-dependent inotropy in cardiomyocytes. Basic Res Cardiol. 2022;117:8. doi: 10.1007/s00395-022-00915-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Winger G, Jutkiewicz EM, Woods JH. Comparison of the muscarinic antagonist effects of scopolamine and L-687,306. Behav Pharmacol. 2020;31:359–367. doi: 10.1097/FBP.0000000000000537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Butova X, Myachina T, Simonova R, Kochurova A, Bozhko Y, Arkhipov M, Solovyova O, Kopylova G, Shchepkin D, Khokhlova Peculiarities of the acetylcholine action on the contractile function of cardiomyocytes from the left and right atria in rats. Cells. 2022;11:3809. doi: 10.3390/cells11233809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Baine S, Thomas J, Bonilla I, Ivanova M, Belevych A, Li J, Veeraraghavan R, Radwanski PB, Carnes C, Gyorke S. Muscarinic-dependent phosphorylation of the cardiac ryanodine receptor by protein kinase G is mediated by PI3K-AKT-nNOS signaling. J Biol Chem. 2020;295:11720–11728. doi: 10.1074/jbc.RA120.014054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ho HT, Belevych AE, Liu B, Bonilla IM, Radwański PB, Kubasov IV, Valdivia HH, Schober K, Carnes CA, Györke S. Muscarinic stimulation facilitates sarcoplasmic reticulum Ca release by modulating ryanodine receptor 2 phosphorylation through protein kinase G and Ca/calmodulin-dependent protein kinase II. Hypertension. 2016;68:1171–1178. doi: 10.1161/HYPERTENSIONAHA.116.07666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cassambai S, Mee CJ, Renshaw D, Hussain A. Tiotropium bromide, a long acting muscarinic receptor antagonist triggers intracellular calcium signalling in the heart. Toxicol Appl Pharmacol. 2019;384:114778. doi: 10.1016/j.taap.2019.114778. [DOI] [PubMed] [Google Scholar]
  • 19.Dolejší E, Janoušková A, Jakubík J. Muscarinic receptors in cardioprotection and vascular tone regulation. Physiological research. 2024 doi: 10.33549/physiolres.935270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Perera RK, Fischer TH, Wagner M, Dewenter M, Vettel C, Bork NI, Maier LS, Conti M, Wess J, El-Armouche A, et al. Atropine augments cardiac contractility by inhibiting cAMP-specific phosphodiesterase type 4. Sci Rep. 2017;7:15222. doi: 10.1038/s41598-017-15632-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kim HY, Choi HR, Lee YJ, Cui HZ, Jin SN, Cho KW, Kang DG, Lee HS. Accentuation of ursolic acid on muscarinic receptor-induced ANP secretion in beating rabbit atria. Life Sci. 2014;94:145–150. doi: 10.1016/j.lfs.2013.12.001. [DOI] [PubMed] [Google Scholar]
  • 22.Kawada T, Sonobe T, Nishikawa T, Hayama Y, Li M, Zheng C, Uemura K, Akiyama T, Pearson JT, Sugimachi M. Contribution of afferent pathway to vagal nerve stimulation-induced myocardial interstitial acetylcholine release in rats. Am J Physiol Regul Integr Comp Physiol. 2020;319:R517–R525. doi: 10.1152/ajpregu.00080.2020. [DOI] [PubMed] [Google Scholar]
  • 23.Bencze M, Boros A, Behuliak M, Vavrinova A, Vaneckova I, Zicha J. Changes in cardiovascular autonomic control induced by chronic inhibition of acetylcholinesterase during pyridostigmine or donepezil treatment of spontaneously hypertensive rats. Eur J Pharmacol. 2024;971:176526. doi: 10.1016/j.ejphar.2024.176526. [DOI] [PubMed] [Google Scholar]
  • 24.Harada N, Ochi K, Yaosaka N, Teraoka H, Hiraga T, Iwanaga T, Unno T, Komori S, Yamada M, Kitazawa T. Immunohistochemical and functional studies for M3 muscarinic receptors and cyclo-oxygenase-2 expressed in the mouse atrium. Auton Autacoid Pharmacol. 2012;32:41–52. doi: 10.1111/j.1474-8673.2012.00472.x. [DOI] [PubMed] [Google Scholar]
  • 25.Stavrakis S, Kem DC, Patterson E, Lozano P, Huang S, Szabo B, Cunningham MW, Lazzara R, Yu X. Opposing cardiac effects of autoantibody activation of β-adrenergic and M2 muscarinic receptors in cardiac-related diseases. Int J Cardiol. 2011;148:331–336. doi: 10.1016/j.ijcard.2009.11.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Camara H, da Silva Junior ED, Garcia AG, Jurkiewicz A, Rodrigues JQD. Cardiac arrest induced by muscarinic or adenosine receptors agonists is reversed by DPCPX through double mechanism. Eur J Pharmacol. 2018;819:9–15. doi: 10.1016/j.ejphar.2017.09.030. [DOI] [PubMed] [Google Scholar]
  • 27.Sassu E, Tumlinson G, Stefanovska D, Fernández MC, Iaconianni P, Madl J, Brennan TA, Koch M, Cameron BA, Preissl S, et al. Age-related structural and functional changes of the intracardiac nervous system. J Mol Cell Cardiol. 2024;187:1–14. doi: 10.1016/j.yjmcc.2023.12.002. [DOI] [PubMed] [Google Scholar]
  • 28.Poller U, Nedelka G, Radke J, Pönicke K, Brodde OE. Age-dependent changes in cardiac muscarinic receptor function in healthy volunteers. J Am Coll Cardiol. 1997;29:187–193. doi: 10.1016/S0735-1097(96)00437-8. [DOI] [PubMed] [Google Scholar]
  • 29.Wang S, Jiang Y, Chen J, Dai C, Liu D, Pan W, Wang L, Fasae MB, Sun L, Wang L, Liu Y. Activation of M3 muscarinic acetylcholine receptors delayed cardiac aging by inhibiting the caspase-1/IL-1beta signaling pathway. Cell Physiol Biochem. 2018;49:1208–1216. doi: 10.1159/000493332. [DOI] [PubMed] [Google Scholar]
  • 30.Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, Himmelfarb CD, Khera A, Lloyd-Jones D, McEvoy JW, et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: A report of the american college of Cardiology/American heart association task force on clinical practice guidelines. Circulation. 2019;140:e596–e646. doi: 10.1161/CIR.0000000000000678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Tompkins JD, Buckley U, Salavatian S, Shivkumar K, Ardell JL. Vagally-mediated heart block after myocardial infarction associated with plasticity of epicardial neurons controlling the atrioventricular node. Front Synaptic Neurosci. 2022;14:960458. doi: 10.3389/fnsyn.2022.960458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Singh S, Loke YK, Furberg CD. Inhaled anticholinergics and risk of major adverse cardiovascular events in patients with chronic obstructive pulmonary disease: A systematic review and meta-analysis. JAMA. 2008;300:1439–1450. doi: 10.1001/jama.300.12.1439. [DOI] [PubMed] [Google Scholar]
  • 33.Mazzadi AN, Pineau J, Costes N, Le Bars D, Bonnefoi F, Croisille P, Porcher R, Chevalier P. Muscarinic receptor upregulation in patients with myocardial infarction: A new paradigm. Circ Cardiovasc Imaging. 2009;2:365–372. doi: 10.1161/CIRCIMAGING.108.822106. [DOI] [PubMed] [Google Scholar]
  • 34.Buchholz B, Kelly J, Munoz M, Bernatené EA, Méndez Diodati N, González Maglio DH, Dominici FP, Gelpi RJ. Vagal stimulation mimics preconditioning and postconditioning of ischemic myocardium in mice by activating different protection mechanisms. Am J Physiol Heart Circ Physiol. 2018;314:H1289–H1297. doi: 10.1152/ajpheart.00286.2017. [DOI] [PubMed] [Google Scholar]
  • 35.Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, Liu Y, Zheng W, Shang H, Zhang J, et al. CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med. 2016;22:175–182. doi: 10.1038/nm.4017. [DOI] [PubMed] [Google Scholar]
  • 36.Lauro FV, Maria LR, Tomas LG, Francisco DC, Rolando GM, Marcela RN, Virginia MA, Alejandra GE, Yazmin OA. Design and synthesis of two new steroid derivatives with biological activity on heart failure via the M2-muscarinic receptor activation. Steroids. 2020;158:108620. doi: 10.1016/j.steroids.2020.108620. [DOI] [PubMed] [Google Scholar]
  • 37.Rinaldi R, Colucci M, Torre I, Ausiello D, Bonanni A, Basile M, Salzillo C, Sanna T, Liuzzo G, Leone AM, et al. Predicting the response to acetylcholine in ischemia or infarction with non-obstructive coronary arteries: The ABCD score. Atherosclerosis. 2024;391:117503. doi: 10.1016/j.atherosclerosis.2024.117503. [DOI] [PubMed] [Google Scholar]
  • 38.Liao F, Zheng Y, Cai J, Fan J, Wang J, Yang J, Cui Q, Xu G, Tang C, Geng B, et al. Catestatin attenuates endoplasmic reticulum induced cell apoptosis by activation type 2 muscarinic acetylcholine receptor in cardiac ischemia/reperfusion. Sci Rep. 2015;5:16590. doi: 10.1038/srep16590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kakinuma Y, Tsuda M, Okazaki K, Akiyama T, Arikawa M, Noguchi T, Sato T. Heart-specific overexpression of choline acetyltransferase gene protects murine heart against ischemia through hypoxia-inducible factor-1α-related defense mechanisms. J Am Heart Assoc. 2013;2:e004887. doi: 10.1161/JAHA.112.004887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Xue RQ, Zhao M, Wu Q, Yang S, Cui YL, Yu XJ, Liu J, Zang WJ. Regulation of mitochondrial cristae remodelling by acetylcholine alleviates palmitate-induced cardiomyocyte hypertrophy. Free Radic Biol Med. 2019;145:103–117. doi: 10.1016/j.freeradbiomed.2019.09.025. [DOI] [PubMed] [Google Scholar]
  • 41.Palee S, Apaijai N, Shinlapawittayatorn K, Chattipakorn SC, Chattipakorn N. Acetylcholine attenuates hydrogen peroxide-induced intracellular calcium dyshomeostasis through both muscarinic and nicotinic receptors in cardiomyocytes. Cell Physiol Biochem. 2016;39:341–349. doi: 10.1159/000445628. [DOI] [PubMed] [Google Scholar]
  • 42.Lv YX, Zhong S, Tang H, Luo B, Chen SJ, Chen L, Zheng F, Zhang L, Wang L, Li XY, et al. VEGF-A and VEGF-B coordinate the arteriogenesis to repair the infarcted heart with vagus nerve stimulation. Cell Physiol Biochem. 2018;48:433–449. doi: 10.1159/000491775. [DOI] [PubMed] [Google Scholar]
  • 43.Travieso A, Jeronimo-Baza A, Faria D, Shabbir A, Mejia-Renteria H, Escaned J. Invasive evaluation of coronary microvascular dysfunction. J Nucl Cardiol. 2022;29:2474–2486. doi: 10.1007/s12350-022-02997-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Alves-Lopes R, Neves KB, Touyz RM. Muscarinic receptor type-3 in hypertension and cholinergic-adrenergic crosstalk: Genetic insights and potential for new antihypertensive targets. Can J Cardiol. 2019;35:555–557. doi: 10.1016/j.cjca.2019.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Khuanjing T, Palee S, Chattipakorn SC, Chattipakorn N. The effects of acetylcholinesterase inhibitors on the heart in acute myocardial infarction and heart failure: From cells to patient reports. Acta Physiol (Oxf) 2020;228:e13396. doi: 10.1111/apha.13396. [DOI] [PubMed] [Google Scholar]
  • 46.Shahim B, Xu H, Haugaa K, Zetterberg H, Jurga J, Religa D, Eriksdotter M. Cholinesterase inhibitors are associated with reduced mortality in patients with Alzheimer's disease and previous myocardial infarction. Eur Heart J Cardiovasc Pharmacother. 2024;10:128–136. doi: 10.1093/ehjcvp/pvad102. [DOI] [PubMed] [Google Scholar]
  • 47.Pan Z, Guo Y, Qi H, Fan K, Wang S, Zhao H, Fan Y, Xie J, Guo F, Hou Y, et al. M3 subtype of muscarinic acetylcholine receptor promotes cardioprotection via the suppression of miR-376b-5p. PLoS One. 2012;7:e32571. doi: 10.1371/journal.pone.0032571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhao J, Su Y, Zhang Y, Pan Z, Yang L, Chen X, Liu Y, Lu Y, Du Z, Yang B. Activation of cardiac muscarinic M3 receptors induces delayed cardioprotection by preserving phosphorylated connexin43 and up-regulating cyclooxygenase-2 expression. Br J Pharmacol. 2010;159:1217–1225. doi: 10.1111/j.1476-5381.2009.00606.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Zhao L, Chen T, Hang P, Li W, Guo J, Pan Y, Du J, Zheng Y, Du Z. Choline attenuates cardiac fibrosis by inhibiting p38MAPK signaling possibly by acting on M3 muscarinic acetylcholine receptor. Front Pharmacol. 2019;10:1386. doi: 10.3389/fphar.2019.01386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Liu H, Hofmann J, Fish I, Schaake B, Eitel K, Bartuschat A, Kaindl J, Rampp H, Banerjee A, Hübner H, et al. Structure-guided development of selective M3 muscarinic acetylcholine receptor antagonists. Proc Natl Acad Sci USA. 2018;115:12046–12050. doi: 10.1073/pnas.1813988115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wang YP, Hang PZ, Sun LH, Zhang Y, Zhao JL, Pan ZW, Ji HR, Wang LA, Bi H, Du ZM. M3 muscarinic acetylcholine receptor is associated with beta-catenin in ventricular myocytes during myocardial infarction in the rat. Clin Exp Pharmacol Physiol. 2009;36:995–1001. doi: 10.1111/j.1440-1681.2009.05176.x. [DOI] [PubMed] [Google Scholar]
  • 52.Harvey KL, Hussain A, Maddock HL. Ipratropium bromide-mediated myocardial injury in in vitro models of myocardial Ischaemia/reperfusion. Toxicol Sci. 2014;138:457–467. doi: 10.1093/toxsci/kfu001. [DOI] [PubMed] [Google Scholar]
  • 53.Nuntaphum W, Pongkan W, Wongjaikam S, Thummasorn S, Tanajak P, Khamseekaew J, Intachai K, Chattipakorn SC, Chattipakorn N, Shinlapawittayatorn K. Vagus nerve stimulation exerts cardioprotection against myocardial ischemia/reperfusion injury predominantly through its efferent vagal fibers. Basic Res Cardiol. 2018;113:22. doi: 10.1007/s00395-018-0683-0. [DOI] [PubMed] [Google Scholar]
  • 54.Pickard JMJ, Burke N, Davidson SM, Yellon DM. Intrinsic cardiac ganglia and acetylcholine are important in the mechanism of ischaemic preconditioning. Basic Res Cardiol. 2017;112:11. doi: 10.1007/s00395-017-0601-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Khuanjing T, Palee S, Kerdphoo S, Jaiwongkam T, Anomasiri A, Chattipakorn SC, Chattipakorn N. Donepezil attenuated cardiac ischemia/reperfusion injury through balancing mitochondrial dynamics, mitophagy, and autophagy. Transl Res. 2021;230:82–97. doi: 10.1016/j.trsl.2020.10.010. [DOI] [PubMed] [Google Scholar]
  • 56.Intachai K, Chattipakorn SC, Chattipakorn N, Shinlapawittayatorn K. Acetylcholine exerts cytoprotection against hypoxia/reoxygenation-induced apoptosis, autophagy and mitochondrial impairment through both muscarinic and nicotinic receptors. Apoptosis. 2022;27:233–245. doi: 10.1007/s10495-022-01715-2. [DOI] [PubMed] [Google Scholar]
  • 57.Xu M, Bi X, He X, Yu X, Zhao M, Zang W. Inhibition of the mitochondrial unfolded protein response by acetylcholine alleviated hypoxia/reoxygenation-induced apoptosis of endothelial cells. Cell Cycle. 2016;15:1331–1343. doi: 10.1080/15384101.2016.1160985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Xue RQ, Sun L, Yu XJ, Li DL, Zang WJ. Vagal nerve stimulation improves mitochondrial dynamics via an M3 receptor/CaMKKbeta/AMPK pathway in isoproterenol-induced myocardial ischaemia. J Cell Mol Med. 2017;21:58–71. doi: 10.1111/jcmm.12938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Li W, Yu J, Yang Y, Wang J, Liu Y, Wang J, Hu J, Yuan Y, Du Z. M3 subtype of muscarinic acetylcholine receptor inhibits cardiac fibrosis via targeting microRNA-29b/beta-site app cleaving enzyme 1 axis. Cardiovasc Diagn Ther. 2024;14:143–157. doi: 10.21037/cdt-23-309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Liu JJ, Huang N, Lu Y, Zhao M, Yu XJ, Yang Y, Yang YH, Zang WJ. Improving vagal activity ameliorates cardiac fibrosis induced by angiotensin II: in vivo and in vitro. Sci Rep. 2015;5 doi: 10.1038/srep17108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Gurses KM, Yalcin MU, Kocyigit D, Kesikli SA, Canpolat U, Yorgun H, Sahiner ML, Kaya EB, Hazirolan T, Ozer N, et al. M2-muscarinic acetylcholine receptor autoantibody levels predict left atrial fibrosis severity in paroxysmal lone atrial fibrillation patients undergoing cryoablation. Europace. 2014;17:239–246. doi: 10.1093/europace/euu228. [DOI] [PubMed] [Google Scholar]
  • 62.Ma G, Wu X, Zeng L, Jin J, Liu X, Zhang J, Zhang L. Association of autoantibodies against M2-muscarinic acetylcholine receptor with atrial fibrosis in atrial fibrillation patients. Cardiol Res Pract. 2019;2019:8271871. doi: 10.1155/2019/8271871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Heijman J, Kirchner D, Kunze F, Chrétien EM, Michel-Reher MB, Voigt N, Knaut M, Michel MC, Ravens U, Dobrev D. Muscarinic type-1 receptors contribute to IK,ACh in human atrial cardiomyocytes and are upregulated in patients with chronic atrial fibrillation. Int J Cardiol. 2018;255:61–68. doi: 10.1016/j.ijcard.2017.12.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Garcia-Domingo M, Garcia-Pedraza JA, Fernandez-Gonzalez JF, Lopez C, Martin ML, Moran A. Fluoxetine treatment decreases cardiac vagal input and alters the serotonergic modulation of the parasympathetic outflow in diabetic rats. Int J Mol Sci. 2022;23:5736. doi: 10.3390/ijms23105736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Jungen C, Scherschel K, Eickholt C, Kuklik P, Klatt N, Bork N, Salzbrunn T, Alken F, Angendohr S, Klene C, et al. Disruption of cardiac cholinergic neurons enhances susceptibility to ventricular arrhythmias. Nat Commun. 2017;8:14155. doi: 10.1038/ncomms14155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gergs U, Wackerhagen S, Fuhrmann T, Schafer I, Neumann J. Further investigations on the influence of protein phosphatases on the signaling of muscarinic receptors in the atria of mouse hearts. Naunyn Schmiedebergs Arch Pharmacol. 2024;39:5731–5743. doi: 10.1007/s00210-024-02973-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Magyar T, Árpádffy-Lovas T, Pászti B, Tóth N, Szlovák J, Gazdag P, Kohajda Z, Gyökeres A, Györe B, Gurabi Z, et al. Muscarinic agonists inhibit the ATP-dependent potassium current and suppress the ventricle-Purkinje action potential dispersion. Can J Physiol Pharmacol. 2021;99:247–253. doi: 10.1139/cjpp-2020-0408. [DOI] [PubMed] [Google Scholar]
  • 68.Voigt N, Friedrich A, Bock M, Wettwer E, Christ T, Knaut M, Strasser RH, Ravens U, Dobrev D. Differential phosphorylation-dependent regulation of constitutively active and muscarinic receptor-activated IK,ACh channels in patients with chronic atrial fibrillation. Cardiovasc Res. 2007;74:426–437. doi: 10.1016/j.cardiores.2007.02.009. [DOI] [PubMed] [Google Scholar]
  • 69.Petersen J, Castro L, Bengaard AKP, Pecha S, Ismaili D, Schulz C, Sahni J, Steenpass A, Meier C, Reichenspurner H, et al. Muscarinic receptor activation reduces force and arrhythmias in human atria independent of IK,ACh. J Cardiovasc Pharmacol. 2022;79:678–686. doi: 10.1097/FJC.0000000000001237. [DOI] [PubMed] [Google Scholar]
  • 70.Couselo-Seijas M, Lopez-Canoa JN, Agra-Bermejo RM, Díaz-Rodriguez E, Fernandez AL, Martinez-Cereijo JM, Durán-Muñoz D, Bravo SB, Velo A, González-Melchor L, et al. Cholinergic activity regulates the secretome of epicardial adipose tissue: Association with atrial fibrillation. J Cell Physiol. 2019;234:10512–10522. doi: 10.1002/jcp.27723. [DOI] [PubMed] [Google Scholar]
  • 71.Deng J, Guo Y, Zhang G, Zhang L, Kem D, Yu X, Jiang H, Li H. M2 muscarinic autoantibodies and thyroid hormone promote susceptibility to atrial fibrillation and sinus tachycardia in an autoimmune rabbit model. Exp Physiol. 2021;106:882–890. doi: 10.1113/EP089284. [DOI] [PubMed] [Google Scholar]
  • 72.Moss R, Sachse FB, Moreno-Galindo EG, Navarro-Polanco RA, Tristani-Firouzi M, Seemann G. Modeling effects of voltage dependent properties of the cardiac muscarinic receptor on human sinus node function. PLoS Comput Biol. 2018;14:e1006438. doi: 10.1371/journal.pcbi.1006438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Liu Y, Sun L, Pan Z, Bai Y, Wang N, Zhao J, Xu C, Li Z, Li B, Du Z, et al. Overexpression of M3 muscarinic receptor is a novel strategy for preventing sudden cardiac death in transgenic mice. Mol Med. 2011;17:1179–1187. doi: 10.2119/molmed.2011.00093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Olivas A, Gardner RT, Wang L, Ripplinger CM, Woodward WR, Habecker BA. Myocardial infarction causes transient cholinergic transdifferentiation of cardiac sympathetic nerves via gp130. J Neurosci. 2016;36:479–488. doi: 10.1523/JNEUROSCI.3556-15.2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Prado MB, Jr, Adiao KJ. Acetylcholinesterase inhibitors in myasthenic crisis: A systematic review of observational studies. Neurocrit Care. 2021;35:528–544. doi: 10.1007/s12028-021-01259-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bober SL, Ciriello J, Jones DL. Atrial arrhythmias and autonomic dysfunction in rats exposed to chronic intermittent hypoxia. Am J Physiol Heart Circ Physiol. 2018;314:H1160–H1168. doi: 10.1152/ajpheart.00173.2017. [DOI] [PubMed] [Google Scholar]
  • 77.Cavalcante GL, Brognara F, Oliveira LVC, Lataro RM, Durand MT, de Oliveira AP, da Nóbrega ACL, Salgado HC, Sabino JPJ. Benefits of pharmacological and electrical cholinergic stimulation in hypertension and heart failure. Acta Physiol (Oxf) 2021;232:e13663. doi: 10.1111/apha.13663. [DOI] [PubMed] [Google Scholar]
  • 78.Baine S, Bonilla I, Belevych A, Stepanov A, Dorn LE, Terentyeva R, Terentyev D, Accornero F, Carnes CA, Gyorke S. Pyridostigmine improves cardiac function and rhythmicity through RyR2 stabilization and inhibition of STIM1-mediated calcium entry in heart failure. J Cell Mol Med. 2021;25:4637–4648. doi: 10.1111/jcmm.16356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Fernandez SF, Canty JM., Jr Adrenergic and cholinergic plasticity in heart failure. Circ Res. 2015;116:1639–1642. doi: 10.1161/CIRCRESAHA.115.306439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Teixeira VP, Miranda K, Scalzo S, Rocha-Resende C, Silva MM, Tezini GCSV, Melo MB, Souza-Neto FP, Silva KSC, Jesus ICG, et al. Increased cholinergic activity under conditions of low estrogen leads to adverse cardiac remodeling. Am J Physiol Cell Physiol. 2021;320:C602–C612. doi: 10.1152/ajpcell.00142.2020. [DOI] [PubMed] [Google Scholar]
  • 81.Ma G, Chen L, Yue Y, Liu X, Wang Y, Shi C, Song F, Shi W, Lo Y, Zhang L. Impact of autoantibodies against the M2-muscarinic acetylcholine receptor on clinical outcomes in peripartum cardiomyopathy patients with standard treatment. BMC Cardiovasc Disord. 2021;21:619. doi: 10.1186/s12872-021-02414-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Li M, Zheng C, Kawada T, Inagaki M, Uemura K, Sugimachi M. Intracerebroventricular infusion of donepezil prevents cardiac remodeling and improves the prognosis of chronic heart failure rats. J Physiol Sci. 2020;70:11. doi: 10.1186/s12576-020-00739-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Williams AM, Shave RE, Coulson JM, White H, Rosser-Stanford B, Eves ND. Influence of vagal control on sex-related differences in left ventricular mechanics and hemodynamics. Am J Physiol Heart Circ Physiol. 2018;315:H687–H698. doi: 10.1152/ajpheart.00733.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Schultheiss HP, Fairweather D, Caforio ALP, Escher F, Hershberger RE, Lipshultz SE, Liu PP, Matsumori A, Mazzanti A, McMurray J, Priori SG. Dilated cardiomyopathy. Nat Rev Dis Primers. 2019;5:32. doi: 10.1038/s41572-019-0084-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chen X, Bai Y, Sun H, Su Z, Guo J, Sun C, Du Z. Overexpression of m3 muscarinic receptor suppressed adverse electrical remodeling in hypertrophic myocardium via increasing repolarizing K+ currents. Cell Physiol Biochem. 2017;43:915–925. doi: 10.1159/000481642. [DOI] [PubMed] [Google Scholar]
  • 86.Wang S, Han HM, Pan ZW, Hang PZ, Sun LH, Jiang YN, Song HX, Du ZM, Liu Y. Choline inhibits angiotensin II-induced cardiac hypertrophy by intracellular calcium signal and p38 MAPK pathway. Naunyn Schmiedebergs Arch Pharmacol. 2012;385:823–831. doi: 10.1007/s00210-012-0740-4. [DOI] [PubMed] [Google Scholar]
  • 87.Ma M, Chen W, Hua Y, Jia H, Song Y, Wang Y. Aerobic exercise ameliorates cardiac hypertrophy by regulating mitochondrial quality control and endoplasmic reticulum stress through M2 AChR. J Cell Physiol. 2021;236:6581–6596. doi: 10.1002/jcp.30342. [DOI] [PubMed] [Google Scholar]
  • 88.Matsui S, Fu ML, Hayase M, Katsuda S, Yamaguchi N, Teraoka K, Kurihara T, Takekoshi N. Active immunization of combined beta1-adrenoceptor and M2-muscarinic receptor peptides induces cardiac hypertrophy in rabbits. J Card Fail. 1999;5:246–254. doi: 10.1016/S1071-9164(99)90009-X. [DOI] [PubMed] [Google Scholar]
  • 89.da Silva Gonçalves Bós D, Van Der Bruggen CEE, Kurakula K, Sun XQ, Casali KR, Casali AG, Rol N, Szulcek R, Dos Remedios C, Guignabert C, et al. Contribution of impaired parasympathetic activity to right ventricular dysfunction and pulmonary vascular remodeling in pulmonary arterial hypertension. Circulation. 2018;137:910–924. doi: 10.1161/CIRCULATIONAHA.117.027451. [DOI] [PubMed] [Google Scholar]
  • 90.Minassa VS, Aitken AV, Hott SC, de Sousa GJ, Batista TJ, Gonçalves RCR, Coitinho JB, Paton JFR, Beijamini V, Bissoli NS, Sampaio KN. Intermittent exposure to chlorpyrifos results in cardiac hypertrophy and oxidative stress in rats. Toxicology. 2022;482:153357. doi: 10.1016/j.tox.2022.153357. [DOI] [PubMed] [Google Scholar]
  • 91.Duan X, Liu R, Luo XL, Gao XJ, Hu FH, Guo C, Wang J, Hu XY, Chun YS, Yuan JS, et al. The relationship between β1-adrenergic and M2-muscarinic receptor autoantibodies and hypertrophic cardiomyopathy. Exp Physiol. 2020;105:522–530. doi: 10.1113/EP088263. [DOI] [PubMed] [Google Scholar]
  • 92.Ribeiro KC, Campelo RP, Rodrigues DDRF, Mattos EC, Brandão IT, da Silva CL, Bouskela E, Martinez CG, Kurtenbach E. Immunization with plasmids encoding M2 acetylcholine muscarinic receptor epitopes impairs cardiac function in mice and induces autophagy in the myocardium. Autoimmunity. 2018;51:245–257. doi: 10.1080/08916934.2018.1514389. [DOI] [PubMed] [Google Scholar]
  • 93.Bezerra OC, Franca CM, Rocha JA, Neves GA, Souza PRM, Teixeira Gomes M, Malfitano C, Loleiro TCA, Dourado PM, Llesuy S, et al. Cholinergic stimulation improves oxidative stress and inflammation in experimental myocardial infarction. Sci Rep. 2017;7:13687. doi: 10.1038/s41598-017-14021-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Barboza CA, Fukushima AR, Carrozzi N, Machi JF, Dourado PMM, Mostarda CT, Irigoyen MC, Nathanson L, Morris M, Caperuto EC, Rodrigues B. Cholinergic stimulation by pyridostigmine bromide before myocardial infarction prevent cardiac and autonomic dysfunction. Sci Rep. 2019;9:2481. doi: 10.1038/s41598-019-38841-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Halder N, Lal G. Cholinergic system and its therapeutic importance in inflammation and autoimmunity. Front Immunol. 2021;12:660342. doi: 10.3389/fimmu.2021.660342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Cox MA, Duncan GS, Lin GHY, Steinberg BE, Yu LX, Brenner D, Buckler LN, Elia AJ, Wakeham AC, Nieman B, et al. Choline acetyltransferase-expressing T cells are required to control chronic viral infection. Science. 2019;363:639–644. doi: 10.1126/science.aau9072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Reardon C, Duncan GS, Brüstle A, Brenner D, Tusche MW, Olofsson PS, Rosas-Ballina M, Tracey KJ, Mak TW. Lymphocyte-derived ACh regulates local innate but not adaptive immunity. Proc Natl Acad Sci USA. 2013;110:1410–1415. doi: 10.1073/pnas.1221655110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.De-Pu Z, Li-Sha G, Guang-Yi C, Xiaohong G, Chao X, Cheng Z, Wen-Wu Z, Jia L, Jia-Feng L, Maoping C, Yue-Chun L. The cholinergic anti-inflammatory pathway ameliorates acute viral myocarditis in mice by regulating CD4+ T cell differentiation. Virulence. 2018;9:1364–1376. doi: 10.1080/21505594.2018.1482179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wang Y, Liu Y, Li XY, Yao LY, Mbadhi M, Chen SJ, Lv YX, Bao X, Chen L, Chen SY, et al. Vagus nerve stimulation-induced stromal cell-derived factor-l alpha participates in angiogenesis and repair of infarcted hearts. ESC Heart Fail. 2023;10:3311–3329. doi: 10.1002/ehf2.14475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Albano GD, Bonanno A, Moscato M, Anzalone G, Di Sano C, Riccobono L, Wenzel SE, Profita M. Crosstalk between mAChRM3 and beta2AR, via acetylcholine PI3/PKC/PBEP1/Raf-1 MEK1/2/ERK1/2 pathway activation, in human bronchial epithelial cells after Long-term cigarette smoke exposure. Life Sci. 2018;192:99–109. doi: 10.1016/j.lfs.2017.11.034. [DOI] [PubMed] [Google Scholar]
  • 101.Wu Q, Zhao M, Li D, He X, Zang W. Cholinergic drugs reduce metabolic inflammation and diabetic myocardial injury by regulating the gut bacterial component lipopolysaccharide-induced ERK/Egr-1 pathway. FASEB J. 2023;37:e22917. doi: 10.1096/fj.202202108R. [DOI] [PubMed] [Google Scholar]
  • 102.Jiang W, Li D, Han R, Zhang C, Jin WN, Wood K, Liu Q, Shi FD, Hao J. Acetylcholine-producing NK cells attenuate CNS inflammation via modulation of infiltrating monocytes/macrophages. Proc Natl Acad Sci USA. 2017;114:E6202–E6211. doi: 10.1073/pnas.1705491114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rocha-Resende C, da Silva AM, Prado MAM, Guatimosim S. Protective and anti-inflammatory effects of acetylcholine in the heart. Am J Physiol Cell Physiol. 2021;320:C155–C161. doi: 10.1152/ajpcell.00315.2020. [DOI] [PubMed] [Google Scholar]
  • 104.Plaschke K, Do TQM, Uhle F, Brenner T, Weigand MA, Kopitz J. Ablation of the right cardiac vagus nerve reduces acetylcholine content without changing the inflammatory response during endotoxemia. Int J Mol Sci. 2018;19:442. doi: 10.3390/ijms19020442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Tarnawski L, Shavva VS, Kort EJ, Zhuge Z, Nilsson I, Gallina AL, Martínez-Enguita D, Heller Sahlgren B, Weiland M, Caravaca AS, et al. Cholinergic regulation of vascular endothelial function by human ChAT+ T cells. Proc Natl Acad Sci USA. 2023;120:e2212476120. doi: 10.1073/pnas.2212476120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Suissa S, Dell'Aniello S, Ernst P. Long-acting bronchodilator initiation in COPD and the risk of adverse cardiopulmonary events: A population-based comparative safety study. Chest. 2017;151:60–67. doi: 10.1016/j.chest.2016.08.001. [DOI] [PubMed] [Google Scholar]
  • 107.Rogliani P, Calzetta L, Matera MG, di Daniele N, Girolami A, Cazzola M, Ora J. Inhaled therapies and cardiovascular risk in patients with chronic obstructive pulmonary disease. Expert Opin Pharmacother. 2019;20:737–750. doi: 10.1080/14656566.2019.1570133. [DOI] [PubMed] [Google Scholar]
  • 108.Shin J, Lee JH. Effects of tiotropium on the risk of coronary heart disease in patients with COPD: A nationwide cohort study. Sci Rep. 2022;12:16674. doi: 10.1038/s41598-022-21038-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Parkin L, Williams S, Sharples K, Barson D, Horsburgh S, Jackson R, Wu B, Dummer J. Dual versus single long-acting bronchodilator use could raise acute coronary syndrome risk by over 50%: A Population-based nested Case-control study. J Intern Med. 2021;290:1028–1038. doi: 10.1111/joim.13348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Arana A, Margulis AV, McQuay LJ, Ziemiecki R, Bartsch JL, Rothman KJ, Franks B, D'Silva M, Appenteng K, Varas-Lorenzo C, Perez-Gutthann S. Variation in cardiovascular risk related to individual antimuscarinic drugs used to treat overactive bladder: A UK cohort study. Pharmacotherapy. 2018;38:628–637. doi: 10.1002/phar.2121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Guo F, Wang Y, Wang J, Liu Z, Lai Y, Zhou Z, Liu Z, Zhou Y, Xu X, Li Z, et al. Choline potects the heart from doxorubicin-induced cardiotoxicity through vagal activation and Nrf2/HO-1 pathway. Oxid Med Cell Longev. 2022;2022:4740931. doi: 10.1155/2022/4740931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Khuanjing T, Ongnok B, Maneechote C, Siri-Angkul N, Prathumsap N, Arinno A, Chunchai T, Arunsak B, Chattipakorn SC, Chattipakorn N. Acetylcholinesterase inhibitor ameliorates doxorubicin-induced cardiotoxicity through reducing RIP1-mediated necroptosis. Pharmacol Res. 2021;173:105882. doi: 10.1016/j.phrs.2021.105882. [DOI] [PubMed] [Google Scholar]
  • 113.Khalaf HA, El-Mansy AAE. The possible alleviating effect of saffron on chlorpyrifos experimentally induced cardiotoxicity: Histological, immunohistochemical and biochemical study. Acta Histochem. 2019;121:472–483. doi: 10.1016/j.acthis.2019.03.003. [DOI] [PubMed] [Google Scholar]
  • 114.Prathumsap N, Ongnok B, Khuanjing T, Arinno A, Maneechote C, Apaijai N, Chunchai T, Arunsak B, Kerdphoo S, Janjek S, et al. Vagus nerve stimulation exerts cardioprotection against doxorubicin-induced cardiotoxicity through inhibition of programmed cell death pathways. Cell Mol Life Sci. 2022;80:21. doi: 10.1007/s00018-022-04678-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wu Q, Zhao M, He X, Xue R, Li D, Yu X, Wang S, Zang W. Acetylcholine reduces palmitate-induced cardiomyocyte apoptosis by promoting lipid droplet lipolysis and perilipin 5-mediated lipid droplet-mitochondria interaction. Cell Cycle. 2021;20:1890–1906. doi: 10.1080/15384101.2021.1965734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Pedzinska-Betiuk A, Weresa J, Schlicker E, Harasim-Symbor E, Toczek M, Kasacka I, Gajo B, Malinowska B. Chronic cannabidiol treatment reduces the carbachol-induced coronary constriction and left ventricular cardiomyocyte width of the isolated hypertensive rat heart. Toxicol Appl Pharmacol. 2021;411:115368. doi: 10.1016/j.taap.2020.115368. [DOI] [PubMed] [Google Scholar]
  • 117.Pickett MA, Dush MK, Nascone-Yoder NM. Acetylcholinesterase plays a non-neuronal, non-esterase role in organogenesis. Development. 2017;144:2764–2770. doi: 10.1242/dev.149831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Wessler I, Kirkpatrick CJ. Acetylcholine beyond neurons: The non-neuronal cholinergic system in humans. Br J Pharmacol. 2008;154:1558–1571. doi: 10.1038/bjp.2008.185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Huang D, Zhang L, Liu Y, Wang J, Zhang J, Baines KJ, Liu G, Hsu AC, Wang F, Chen Z, et al. Activated non-neuronal cholinergic system correlates with non-type 2 inflammation and exacerbations in severe asthma. Ann Allergy Asthma Immunol. 2024;133:e64–e72.e4. doi: 10.1016/j.anai.2024.03.009. [DOI] [PubMed] [Google Scholar]
  • 120.Braczko F, Fischl SR, Reinders J, Lieder HR, Kleinbongard P. Activation of the nonneuronal cholinergic cardiac system by hypoxic preconditioning protects isolated adult cardiomyocytes from hypoxia/reoxygenation injury. Am J Physiol HeartCirc Physiol. 2024;327:H70–H79. doi: 10.1152/ajpheart.00211.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Chotirat S, Suriyo T, Hokland M, Hokland P, Satayavivad J, Auewarakul CU. Cholinergic activation enhances retinoic acid-induced differentiation in the human NB-4 acute promyelocytic leukemia cell line. Blood Cells Mol Dis. 2016;59:77–84. doi: 10.1016/j.bcmd.2016.04.009. [DOI] [PubMed] [Google Scholar]
  • 122.Oikawa S, Kai Y, Mano A, Sugama S, Mizoguchi N, Tsuda M, Muramoto K, Kakinuma Y. Potentiating a non-neuronal cardiac cholinergic system reinforces the functional integrity of the blood brain barrier associated with systemic anti-inflammatory responses. Brain Behav Immun. 2019;81:122–137. doi: 10.1016/j.bbi.2019.06.005. [DOI] [PubMed] [Google Scholar]
  • 123.Rocha-Resende C, Weinheimer C, Bajpai G, Adamo L, Matkovich SJ, Schilling J, Barger PM, Lavine KJ, Mann DL. Immunomodulatory role of non-neuronal cholinergic signaling in myocardial injury. JCI Insight. 2019;5:e128961. doi: 10.1172/jci.insight.128961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Kakinuma Y, Akiyama T, Sato T. Cholinoceptive and cholinergic properties of cardiomyocytes involving an amplification mechanism for vagal efferent effects in sparsely innervated ventricular myocardium. FEBS J. 2009;276:5111–5125. doi: 10.1111/j.1742-4658.2009.07208.x. [DOI] [PubMed] [Google Scholar]
  • 125.Oikawa S, Kai Y, Mano A, Ohata H, Nemoto T, Kakinuma Y. Various regulatory modes for circadian rhythmicity and sexual dimorphism in the non-neuronal cardiac cholinergic system. J Cardiovasc Transl Res. 2017;10:411–422. doi: 10.1007/s12265-017-9750-4. [DOI] [PubMed] [Google Scholar]
  • 126.Oikawa S, Kai Y, Mano A, Nakamura S, Kakinuma Y. A novel nitric oxide donor, S-Nitroso-NPivaloyl-D-Penicillamine, activates a non-neuronal cardiac cholinergic system to synthesize acetylcholine and augments cardiac function. Cell Physiol Biochem. 2019;52:922–934. doi: 10.33594/000000064. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from International Journal of Molecular Medicine are provided here courtesy of Spandidos Publications

RESOURCES