927
Views
0
CrossRef citations to date
0
Altmetric
Review

Transient receptor potential vanilloid type 1: cardioprotective effects in diabetic models

, , , &
Article: 2281743 | Received 05 May 2023, Accepted 17 Oct 2023, Published online: 20 Nov 2023

ABSTRACT

Cardiovascular disease, especially heart failure (HF) is the leading cause of death in patients with diabetes. Individuals with diabetes are prone to a special type of cardiomyopathy called diabetic cardiomyopathy (DCM), which cannot be explained by heart diseases such as hypertension or coronary artery disease, and can contribute to HF. Unfortunately, the current treatment strategy for diabetes-related cardiovascular complications is mainly to control blood glucose levels; nonetheless, the improvement of cardiac structure and function is not ideal. The transient receptor potential cation channel subfamily V member 1 (TRPV1), a nonselective cation channel, has been shown to be universally expressed in the cardiovascular system. Increasing evidence has shown that the activation of TRPV1 channel has a potential protective influence on the cardiovascular system. Numerous studies show that activating TRPV1 channels can improve the occurrence and progression of diabetes-related complications, including cardiomyopathy; however, the specific mechanisms and effects are unclear. In this review, we summarize that TRPV1 channel activation plays a protective role in the heart of diabetic models from oxidation/nitrification stress, mitochondrial function, endothelial function, inflammation, and cardiac energy metabolism to inhibit the occurrence and progression of DCM. Therefore, TRPV1 may become a latent target for the prevention and treatment of diabetes-induced cardiovascular complications.

Introduction

Diabetes is a systemic metabolic disease usually associated with hypertension, hyperlipidemia, hyperuricemia, obesity, and heart failure (HF) [Citation1]. Diabetes is considered a risk factor for cardiovascular disease, a major cause of increased mortality among patients with diabetes [Citation2]. Data from China demonstrate that 30% of patients with diabetes experience adverse cardiovascular events, accounting for 20% of all mortalities among this population [Citation3]. Diabetes and HF often coexist and are closely related; diabetes increases the risk of HF, and the evaluation, treatment, and prognosis of patients with HF are worse in the context of diabetes [Citation4]. The epidemiological link between diabetes and HF was confirmed in the 1974 Framingham study. After adjusting for relevant risk factors (such as age and concomitant complications), diabetes increased the incidence of HF by 2.5-fold in men and by 5-fold in women [Citation5]. Diabetes accounts for 1/3 of the total number of patients in clinical studies related to HF and is an independent risk factor for poor prognosis [Citation6–8]. Due to the accelerated aging of the population, diabetes-related HF has become prevalent worldwide [Citation9] and threatens the lives and health of people with diabetes.

Characteristics of a diabetic heart

As early as 1972, the autopsy pathology of four patients found that [Citation10] they showed symptoms of HF, with no evidence to support coronary or valvular disease. This is defined as myocardial structural changes and ventricular dysfunction unrelated to other cardiac risk factors, namely diabetic cardiomyopathy (DCM) [Citation11,Citation12]. Generally, this is a kind of myocardial disease that occurs in patients with diabetes and cannot be attributed to conditions like hypertension, coronary atherosclerotic heart disease and other heart diseases. Patients often have clinical manifestations such as angina, arrhythmia and even HF. In an experimental animal model of diabetes, it was also found that diastolic and systolic functions of the heart decreased, accompanied by a decrease in the contraction of cardiomyocytes and variations in the level of specific cardiomyocyte proteins [Citation13,Citation14]. Later studies further found that DCM is characterized by 1) increased myocardial fibrosis and hardness and 2) increased atrial enlargement and left ventricular end-diastolic pressure [Citation15]. The exact cause of diabetes-related cardiac dysfunction is unclear. Still, many basic contributing factors have been reported, including cardiac fibrosis, cardiac hypertrophy, endothelial dysfunction, oxidative stress, inflammation, and mitochondrial dysfunction [Citation16] (). The increased apoptosis and autophagy damage caused by these disorders are also related to cardiac remodeling in DCM [Citation17].

Figure 1. Basic mechanism of diabetes cardiomyopathy. Systemic hyperglycemia, insulin resistance, and deposition of advanced glycation end products (AGEs) caused by diabetes can induce cardiac metabolic changes, promote myocyte inflammation, endothelial cell damage, and mitochondrial dysfunction. These pathways interact with each other, directly or indirectly leading to cardiac hypertrophy, fibrosis, and ischemia, ultimately leading to dysfunction of cardiac relaxation and contraction.

Figure 1. Basic mechanism of diabetes cardiomyopathy. Systemic hyperglycemia, insulin resistance, and deposition of advanced glycation end products (AGEs) caused by diabetes can induce cardiac metabolic changes, promote myocyte inflammation, endothelial cell damage, and mitochondrial dysfunction. These pathways interact with each other, directly or indirectly leading to cardiac hypertrophy, fibrosis, and ischemia, ultimately leading to dysfunction of cardiac relaxation and contraction.

Cardiac insufficiency in patients with diabetes is usually asymptomatic and subclinical in the early stages and is not detected until the terminal stage [Citation18]. It then develops into severe diastolic dysfunction with a normal ejection fraction and eventually into systolic dysfunction in clinical HF with a decreased ejection fraction [Citation11]. Even in asymptomatic patients with good blood glucose control, about 50% showed some degree of cardiac insufficiency [Citation19,Citation20]. Therefore, early diagnosis and treatment of diabetic cardiac complications are required. The current treatment strategy focuses on blood sugar regulation. At the same time, there is a lack of specific treatment and effective clinical management for the diabetic heart, although the sodium-glucose cotransporter-2 inhibition [SGLT2i] offers some hope in this regard [Citation21,Citation22]. Therefore, the need for effective prevention and treatment of cardiovascular complications related to diabetes is important.

Transient receptor potential vanilloid type 1

The transient receptor potential cation channel, subfamily V member 1 (TRPV1), is a nonselective cation channel with obvious permeability to ions, such as H+, Na+, Ca2+, and Mg2+ [Citation23]. The TRPV1 channel comprises four subunits, each containing six transmembrane helices [Citation24]. TRPV1 channels exist as tetramers with C and N terminals facing the cytoplasmic surface [Citation25]. The channel can be activated by a variety of physical or chemical substances, such as temperatures (>42°C), low pH, capsaicin, arachidonic acid, bradykinin, cannabinoids, and free radical metabolites [Citation26,Citation27].

TRPV1 channels are widely distributed in various organs, such as the lung, liver, kidney, intestine, and brain [Citation28], as well as in the cardiovascular system, such as cardiomyocytes [Citation29], epicardium [Citation30], sensory nerve fibers innervating the myocardium [Citation31], vascular endothelial cells, and smooth muscle cells () [Citation28]. However, some studies have demonstrated no expression of TRPV1 in cardiomyocytes [Citation32,Citation33]. Such differences may be owing to different sensitivities of antibodies used in immunocytochemical approaches or inappropriate signal to noise ratio. Additionally, recent data show that the sensitivity of TRPV1 channels to agonist activation can be regulated by TRPA1 channel agonists [Citation34]. When two receptors are co-expressed in the same cell, there is a crosstalk between them. The primary finding of the study is the absence of functional TRPA1 and TRPV1 in murine cardiomyocytes [Citation32]. Further research is required to determine the expression and degree of TRPV1 in cardiomyocytes, as well as whether it experiences interference from other receptor channels.

Table 1. Expression of TRPV1 in Cardiovascular system and its effect on cardiovascular system after activation.

Activation of this channel can lead to the release of neurotransmitters from peripheral nerve endings, including substance P (SP), calcitonin gene-related peptide (CGRP), pituitary adenylate cyclase‐activating polypeptide (PACAP), somatostatin (SST) and other neuropeptides resulting in cardiovascular response [Citation48,Citation49] (). These neuropeptides play crucial roles in the heart both locally (local efferent function) and via the bloodstream (systemic efferent function) [Citation50]. Moreover, CGRP can dilate blood vessels and reduce blood pressure by increasing intracellular NO and K concentrations through endothelial and non-endothelial dependent mechanisms. CGRP can also lower blood pressure by antagonizing the renin-angiotensin-aldosterone system (RAAS) and sympathetic nervous system [Citation51].

Figure 2. Schematic diagram of CGRP and SP release caused by TRPV1 activation. Substance P and CGRP-positive dorsal root ganglia (DRG) neurons often co-express transient receptor potential vanilloid (TRPV1) channel. When TRPV1 is activated, the conformation of TRPV1 changes, the channel opens, the permeability to cations (mainly Ca2+) increases, 1) the influx of cations such as Ca2+ produces depolarizing action potential;2) CGRP is packaged and co-released with substance P in a calcium dependent manner due to depolarization or increased intracellular calcium.

Figure 2. Schematic diagram of CGRP and SP release caused by TRPV1 activation. Substance P and CGRP-positive dorsal root ganglia (DRG) neurons often co-express transient receptor potential vanilloid (TRPV1) channel. When TRPV1 is activated, the conformation of TRPV1 changes, the channel opens, the permeability to cations (mainly Ca2+) increases, 1) the influx of cations such as Ca2+ produces depolarizing action potential;2) CGRP is packaged and co-released with substance P in a calcium dependent manner due to depolarization or increased intracellular calcium.

The potential protective functions of TRPV1 on the cardiovascular system are being increasingly recognized. Earlier studies have found that a variety of neurotransmitters released by TRPV1 can directly protect the heart against damage [Citation52], protect the heart from the adverse effects of ischemic stress [Citation52], improve coronary artery dysfunction, prolong the lifespan of atherosclerotic mice [Citation53], and inhibit oxidative stress caused by high salt-induced hypertension [Citation54].

Our review sums up the cardiovascular effects of TRPV1 in diabetic models, which are believed to be related to the occurrence and development of DCM. Hopefully, this information will help evaluate the cardioprotective effect of TRPV1 in diabetic models.

TRPV1-mediated cardioprotective mechanism in diabetic models

Inhibition of oxidative stress

Oxidative stress is an imbalance between excessive reactive oxygen species (ROS) production and degradation capacity, which tends to be in an oxidative state, leading to inflammatory infiltration in the body and an important factor in cell aging and disease. As the antioxidant content of the normal heart is lower than that of other organs, the heart is extremely vulnerable to oxidative stress and injury [Citation55,Citation56]. Cardiac ROS are mainly derived from uncoupled nitric oxide (NO) synthase, mitochondrial respiratory chain, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, which can be triggered by diversified stimuli, such as tumor necrosis factor-α (TNF-α), angiotensin II (Ang-II) and endothelin-1 (ET-1) [Citation57,Citation58]. In the initial phase of diabetes, multiple antioxidants in the heart are likely to be upregulated to compensate; however, in the late period, diabetes not only produces additional ROS and/or reactive nitrogen species (RNS), but also, more importantly, compromises the antioxidant capacity of the heart [Citation59,Citation60]. Overall, unbridled plasma glucose levels and low-grade systemic inflammation trigger excessive oxidative stress in the cardiovascular system [Citation61]. Increased ROS levels increase the possibility of arrhythmias [Citation62], especially in structurally impaired left ventricular hypertrophic heart disease like DCM [Citation63].

Uncoupling protein 2 (UCP2), a member of the mitochondrial anion carrier family, is a physiological regulator of mitochondrial ROS production and may help prevent diabetes [Citation64,Citation65]. One study demonstrated that oxidative stress induces the expression of UCP2, which prevents the production of ROS. ROS modulates diverse signal trans duction pathways, leading to increased expression of cell adhesion molecules, initiation of pro-inflammatory pathways, activation of matrix metalloproteinase, vascular smooth muscle cell proliferation and death, and endothelial dysfunction and lipid peroxidation – factors implicated in atherogenesis. Therefore, augmented UCP2 activity might restrict ROS production, thereby reducing the hazard of atherosclerosis in patients with diabetes [Citation66]. Studies have indicated that the administration of capsaicin or its analogues can increase the expression of UCP2 in fatty and liver tissues [Citation67,Citation68]. In addition, Sun et al. [Citation69] found that capsaicin could reverse the decrease in TRPV1 expression and protein kinase A (PKA) phosphorylation in endothelial cells induced by high blood sugar and reduce the level of ROS through the PKA/UCP2 pathway. This study further found that compared with normal glucose exposure, high glucose culture impaired endothelium-dependent relaxation of aortic rings in Wild Type (WT) and TRPV1 knockout (TRPV1-) mice. Capsaicin improved endothelium-dependent relaxation of the aortic rings in WT mice but has no effect on TRPV1- mice. We conclude that dietary capsaicin-activated TRPV1 stimulates endothelial PKA phosphorylation, motivates UCP2 expression, inhibits oxidative stress, and promotes endothelial-dependent relaxation in diabetic mice.

Nuclear factor erythroid-related factor 2 (Nrf2) is a transcription factor that regulates the expression of antioxidant genes [Citation70]. In previous studies, Nrf2 not only regulates ROS levels in normal hearts but also plays a part in diabetic hearts [Citation71]. It was further reported that the Nrf2 expression was precipitously downregulated in the hearts of diabetic animals and patients [Citation72]. The downregulation of Nrf2 is an important cause of various diabetic complications [Citation73–75], which may partly explain the occurrence and progression of DCM. It has been found that the influx of Ca2+ produced by TRPV1 activation enhances the phosphorylation of calcium/calmodulin-dependent protein kinase II (CaMKII), which then further upregulates Nrf2 [Citation76]. Therefore, we speculated that TRPV1 may improve oxidative stress in diabetic hearts by upregulating Nrf2 expression.

Alleviating endothelial dysfunction and microvascular injury

Chronic and recurrent hyperglycemia can increase endothelial oxidative metabolism and reduce the bioavailability of NO, an endothelium-derived vasodilator [Citation77]. The endothelium of the arterioles maintains the contractile and diastolic function of the blood vessels through “real-time monitoring” of the blood flow, and further regulates the blood pressure of the body and the pumping blood of the heart. Additionally, endothelium protects body tissues from toxic substances and plays a pivotal role in regulating blood coagulation, maintaining electrolyte balance and facilitating substance transport between tissues and organs. Impairment of these endothelial functions, often leads to vascular dysfunction, which is associated with heart disease. Endothelial dysfunction is inextricably linked to the evolution and degree stratification of cardiovascular diseases in patients with diabetes [Citation78]. In the pathogenesis of decreased NO bioavailability and endothelial dysfunction caused by diabetes and obesity, disturbances in calcium handling in endothelial cells make a critical difference in disturbing the dynamic balance of endothelial cells. Substantial evidence suggests that Transient receptor potential (Trp) channels are involved in calcium regulation in endothelial cells and to be pivotal in vascular dysfunction caused by obesity and diabetes [Citation79].

Li [Citation80] pointed out in a study that model mice with Type 2 Diabetes Mellitus (T2DM) showed 1) decreased expression of TRPV1 in cardiac microvascular endothelial cells (CMECs); 2) inhibition of mitochondrial function, such as the production of ATP and decrease in mitochondrial membrane potential; 3) decreased production of endothelial NO and increased apoptosis of endothelial cells; and 4) destruction of cardiac microvascular integrity, such as irregular valgus and invagination. Impaired diastolic function, such as a decrease in the ratio of the early mitral diastolic wave to the late mitral diastolic wave, showed more severe microvascular damage and diastolic dysfunction in TRPV1- diabetic mice. After supplementation with the OPA1 gene in vitro, the damage of TRPV1- to vascular endothelial cells in diabetes partially improved. Capsaicin noticeably added the mRNA expression of TRPV1, PGC-1a, and OPA1 in the CMEC of WT mice but this phenomenon was not seen in TRPV1- mice. It can be seen that capsaicin activates TRPV1 to reduce the microvascular injury of the diabetic heart through the PGC-1/OPA1 signal.

Additionally, capsaicin-activated TRPV1 can improve the proliferation, migration, and oxidative stress of vascular smooth muscle cells lured by oxidized LDL by upregulating the expression of peroxisome proliferator-activated receptor (PPAR) α and retarding vascular remodeling [Citation81]. Zhu et al. pointed out [Citation82] that the aging of blood vessels is accelerated in patients with diabetes compared to healthy individuals. Capsaicin increases Silent mating type information regulation 2 homolog-1 (SIRT1) levels through the TRPV1/[Ca 2 +] i/CaMKII/AMPK channel. It downregulates the aging marker p21, thus insulating endothelial cells from senescence caused by intermittent hyperglycemia, such as alleviating G0/G1 stagnation, increasing cell viability, and reducing senescent cell counts and ROS production.

Inhibition of inflammation

Inflammation is a self-defense response that occurs when the body is exposed to various damaging factors. Chronic low-grade inflammation is the “common soil” of diabetes and cardiovascular disease. Diabetes is a systemic low-grade inflammatory disease. Hyperglycemia directly induces the secretion and upregulation of cytokines, chemokines, and adhesion molecules in cardiomyocytes by regulating diversified signal pathways, which are based on the nuclear factor kappa-B (NF- κ B) [Citation83–85]. NF-κ B is one of the critical transcription factors regulating pro-inflammatory cytokines, pro-fibrosis genes, and cell survival expression [Citation86]. After these basic molecular events, leukocytes infiltrate the myocardium and increase ROS production by secreting cytokines and fibrogenic factors, thus perpetuating the inflammatory process [Citation83–85]. Additionally, activation of the renin-angiotensin-aldosterone system and advanced glycation end products mediates the aseptic inflammation of cardiomyocytes of patients with diabetes, mainly by acting on Toll-like receptors (TLR) [Citation87–89].

It has been reported that the inhibition of inflammation can improve cardiac insufficiency in diabetic hearts [Citation90]. Still, the absence of TRPV1 can enhance local inflammation and accelerate the attack of systemic inflammatory response syndrome [Citation91]. A study [Citation37] suggested that TRPV1 inhibits the endothelial cells inflammation by activating eNOS, which is characterized by the production of proinflammatory cytokines/chemokines and decreased expression of adhesion molecules. The findings as well showed that TRPV1 induces these effects through activating the Ca2±dependent PI3K/Akt/eNOS/NO signaling pathway and inhibiting NF-κB, which is in accordance with the report of Ching [Citation92]. Furthermore, endovanilloids, a TRPV1 activator, can curb the activation of NF-κB and nuclear factor of activated T cells (NFAT) [Citation93,Citation94]. Huang et al. also pointed out that the deletion of TRPV1 can increase inflammation after myocardial infarction [Citation95].

What’s more, diabetes can promote the tissue infiltration of macrophages and polarize them to M1-like “pro-inflammatory” phenotypes [Citation96,Citation97]. Lv et al. uncovered that TRPV1 inhibits the polarization of M1 macrophages during the progression of osteoarthritis [Citation98]. Zhao et al. certified that the activation of TRPV1 is able to prevent lipid accumulation induced by oxidized LDL and macrophage inflammation induced by TNF-α [Citation99]. Therefore, TRPV1 may alleviate inflammation in the diabetic heart by suppressing the polarization of M1 macrophages.

Improving the function of mitochondria

Of the intracellular ATP in cardiomyocytes, 90% are produced by mitochondrial oxidative phosphorylation; however, in T2DM, mitochondria convert from glucose oxidation to Free Fatty Acids oxidation to produce ATP [Citation18], accompanied by boosted mitochondrial ROS production, damaged oxidative phosphorylation, and calcium overload, resulting in mitochondrial respiratory dysfunction and further autophagy and necrosis of cardiomyocytes [Citation100,Citation101]. Mitochondrial dysfunction is of great importance in the development of DCM and related HF [Citation102].

Luo et al. pointed out [Citation103] that dietary capsaicin upregulates the expression of PGC-1α by activating TRPV1, which increases the quality of mitochondria, improves the respiratory function of mitochondria, and promotes the transformation of glycolysis type II fibers to more oxidizing type I fibers. Previous studies have shown that because type I fibers have more mitochondria, they produce more ATP from lipid oxidation but less ATP from glycolysis than type II fibers, thus offering stabilized energy over a lasting period and reducing lactic acid stockpile [Citation104,Citation105].

In addition, research by Wu et al. found that [Citation106,Citation107] hyperglycemia decreased the activity of AMP-activated protein kinase (AMPK) and triggered the formation of abnormal MAM mitochondrial-associated endoplasmic reticulum (MAM) mediated by Fundc1, leading to mitochondrial dysfunction and cardiomyopathy. Previous studies have shown that Fundc1, a mitochondrial extracellular protein, is of great significance for mitochondrial autophagy and MAMs [Citation107]. Recently, Xiao et al. [Citation108] reported that TRPV1 activation antagonized diabetic nephropathy by reducing Fundc1 transcription and improving mitochondrial function. Therefore, it can be speculated that the activation of TRPV1 can also antagonize DCM by improving Fundc1-related mitochondrial function.

Reduced myocardial fibrosis and cardiac hypertrophy

The earliest autopsy and biopsy evidence showed that an arresting increase in type I and III interstitial collagen deposition in patients with diabetes match non-diabetic myocardial biopsies [Citation109]. Increased expression of TGF-β was reported in diabetic animal models, and this phenomenon was associated with cardiac fibrosis [Citation110–112]. Related studies have found that the TGF-β signal transduction pathway leads to diabetic cardiac fibrosis by inducing cardiomyocyte apoptosis [Citation113], stimulating excessive production of fibroblasts [Citation114], and increasing cardiomyocyte extracellular matrix deposition. The effect of TRPV1 on improving myocardial fibrosis has been widely covered in previous studies. Activation of TRPV1 inhibits cardiac hypertrophy and fibrosis [Citation42,Citation43]. Wang et al. [Citation115] found that TRPV1 activation attenuates the proliferation and differentiation of mouse fibroblasts induced by isoproterenol and prevents the development of myocardial fibrosis. In addition, long-term capsaicin intervention significantly improved myocardial hypertrophy and fibrosis in mice induced by high salt but did not improve in TRPV1- mice [Citation43]. Similarly, continuous treatment of capsaicin reduced myocardial fibrosis in the hearts of diabetic mice but not in TRPV1- mice [Citation80]. It can be seen that the benefits of TRPV1 in improving diabetic cardiac fibrosis is clear, but the specific mechanism remains to be studied.

Activate metabolic regulators and regulate cardiac energy metabolism

Impaired lipid metabolism is an important sign of diabetes, and is related to the occurrence of DCM [Citation116]. The heart derives its main energy from fatty acids instead of glucose as its source. However, in diabetes, the proportion of fatty acid use is higher, on account of insulin resistance and reduced glucose intake [Citation117]. Excessive fatty acid intake over the oxidation rate of the heart, results in an increase in the concentration of active lipids in cardiomyocytes and an increase in ROS production accrue to the β oxidation of concentrated lipids [Citation116]. Several investigations have shown that changes in cardiomyocyte metabolism are of poverty touches of diabetic cardiac dysfunction [Citation118–120]. Although the mechanism of the two remains unclear, it is mainly summarized by several factors, including disruption of calcium homeostasis, reduced cardiac efficiency, lipotoxicity, and myocardial mitochondrial damage. Capsaicin increases fat oxidation, prevents myocardial steatosis, and improves heart and liver function by regulating processes such as adipocyte browning and activating metabolic regulators, including AMPK, PPARα, glucagon-like peptide-1 (GLP-1) and uncoupling protein 1 (UCP1) [Citation121]. These reactions of capsaicin are mediated by the excitation of TRPV1.

Regulating blood glucose and improving insulin sensitivity

TRPV1 receptor is expressed in rat islet β cells and β cell lines and affects insulin secretion [Citation122]. This study also found that capsaicin dose-dependently increased insulin secretion in vitro, and this effect was suppressed by preconditioning with a TRPV1 antagonist. TRPV1 receptors also exist in the afferent sensory nerves innervating the islets. Therefore, TRPV1 activation may regulate insulin secretion directly by changing the secretion of β cells or indirectly by regulating sensory nerves [Citation123]. Razavi et al. have put forward arguments that sensory neurons expressing TRPV1 master islet inflammation and insulin resistance [Citation124]. Zhong et al. speculated that [Citation125] TRPV1 mediates glucose-induced insulin secretion through the release of CGRP and SP. Other researchers have found that TRPV1 receptors exist in intestinal cells expressing GLP-1 and regulate glucose homeostasis and improve insulin sensitivity by inducing intestinal GLP-1 secretion [Citation126,Citation127]. TRPV1 activation regulates blood glucose homeostasis and even enhances insulin sensitivity in many ways.

Conclusion

Currently, hospitalization and mortality rates have significantly increased in patients with diabetes-related HF. Glycemic control effectively minimizes the possibility of HF in patients with diabetes [Citation128]. For every 1% reduction in HbA1C, the risk of HF is reduced by 16% [Citation129]. summarizes the main effects of different types of hypoglycemic drugs on DCM and HF. Nevertheless, hypoglycemic therapy is ineffective in terms of cardiovascular endpoints [Citation16]. Long-term blood glucose control cannot reverse cardiac structural remodeling and dysfunction. In some trials, it has not been proven that the hospitalization rate of HF in diabetic patients treated with antihyperglycemic drugs is lower [Citation128]. Current guidelines for diagnosing and treating diabetes recommend a comprehensive approach to reduce the risk of diabetes-related complications. Special emphasis is placed on managing blood sugar, blood pressure and blood lipids, as well as lifestyle changes, as the basic elements to reduce and delay the cardiovascular risks associated with diabetes [Citation130]. Another strategy to prevent DCM is reducing excessive fatty acid oxidation and lipotoxicity in the heart. Statins have been shown to reduce the incidence and mortality of coronary artery events in patients with diabetes with or without a history of cardiovascular disease [Citation131]. SGLT2 inhibitors can reduce the accumulation of lipids in visceral fat and fat decomposition. The protective effect on myocardial lipotoxicity is not clear. Further research is needed to clarify this point.

Table 2. Summary of the main effects of different glucose-lowering agents on diabetic cardiomyopathy and heart failure.

Recently, GLP-1 agonists and SGLT2 inhibitors have been show to possess cardioprotective properties independent of the hypoglycemic pathway in patients with diabetes [Citation132], as shown in . According to the 2023 American Diabetes Association (ADA) guidelines, regardless of baseline HbA1C levels, for patients with type 2 diabetes who have atherosclerotic heart disease or a high risk of cardiovascular disease or HF, SGLT2 inhibitors and/or GLP-1 agonists are recommended to reduce blood sugar and overall cardiovascular risk, including major cardiovascular adverse events and/or hospitalization risk for HF [Citation130]. However, these two kinds of drugs also have many adverse reactions and limitations. Particularly, SGLT2 inhibitors cause an increased risk of urinary tract infection, lower limb amputation, diabetic ketoacidosis, fracture, and acute renal injury. GLP-1 agonists cause gastrointestinal reactions and an increased risk of pancreatitis and cholelithiasis. Moreover, since GLP-1 agonists is an injection drug, patient’s compliance is relatively poor. GLP-1 agonists and SGLT2 inhibitors offer new hope for preventing and treating diabetes. We speculate that TRPV1 analog also have a protective effect on the heart of diabetes. Further studies are required to clarify the potential targets and related mechanisms of TRPV1 channels in the heart of diabetes.

Table 3. Summary for the effects and adverse reactions of SGLT-2i and GLP-1Ras on diabetes heart.

In summary, based on current studies on patients with diabetes and animal experiments, activating TRPV1 can prevent the occurrence and development of diabetic organ dysfunction through a variety of different mechanisms, such as the inhibition of oxidative stress and inflammation, improvement of mitochondrial function, reduction of endothelial dysfunction and microvascular injury, and reduction of myocardial fibrosis and hypertrophy. However, Mandy et al. reported that 12 (S)-HETE, an effective TRPV1 activator, directly mediates endothelial calcium homeostasis disorders, mitochondrial lesions, and endothelium-dependent vasodilation in patients with diabetes [Citation144]. They further pointed out that eliminating the 12 (S)-HETE/TRPV1 interaction in the TRPV1 Trp box could protect the function of diabetic vascular endothelial cells, increase left ventricular capillary density, and inhibit the worsening of HF with preserved ejection fraction caused by diabetes [Citation145]. It seems that TRPV1 has complex effects under pathophysiological conditions, which could be the upshot of experimental differences. We speculate that different activators, antagonists, and gene knockout techniques may lead to differences in cardiovascular effects on TRPV1. The lipid peroxide 12 (S)-HETE is increased specifically in type 1 diabetic mice and humans and is also an endogenous activator of TRPV1. The plasma concentration of 12 (S)-HETE is particularly high in patients with type 1 diabetes diagnosed for the first time [Citation146] and adult diabetic patients with vascular complications [Citation147,Citation148]. Most current studies use capsaicin to activate the TRPV1 channel to protect the heart from DCM; however, research on other TRPV1 activators or antagonists is not clear and developed enough, and the potency of TRPV1 analogues in the cure of cardiovascular disease is only just emerging. It is also believed that capsaicin has a biphasic effect on the vascular system: at lower concentrations, capsaicin (up to 10 nM) causes vasodilation in the skin due to sensory nerve activation, while higher concentrations (0.1–1 μ M) cause massive contraction in skeletal muscle arterioles due to non-neuronal TRPV1 stimulation [Citation149]. It is not clear whether this difference is due to receptor sensitivity or the difference in TRPV1 receptor density between the two tissues. Further research is needed to clarify the potential targets and related mechanisms of TRPV1 channels in the diabetic heart, to provide medicine professionals unique TRPV1 agonists to prevent and treat diabetes-related cardiovascular diseases.

Thus far, no consensus has been reached on the optimal management strategy for preventing or treating of diabetes-related cardiovascular complications. Current published studies have confirmed the latent protective roles of TRPV1 against atherosclerosis, cardiac ischemic stress, and pathological hypertrophy. Considering that TRPV1 may also have a unique cardioprotective effect in diabetic models, this study summarized the benefits of the TRPV1 channel on blood glucose and cardiovascular diseases in diabetic models. We hope to aid in the diagnosis, prevention, and treatment of cardiovascular diseases in patients with diabetes.

Disclosure statement

The authors do not declare any conflict of interest relevant to this manuscript.

Data availability statement

No data are available for this review.

Additional information

Funding

This study was supported by the National Natural Science Foundation of China under Grant No. 81670447; the Zhejiang Provincial Health Commission Project under Grant No. 2017KY559. LW is sponsored by Zhejiang Provincial Program for the Cultivation of High-Level Innovative Health Talents.

References

  • Fernandez-Twinn DS, Hjort L, Novakovic B, et al. Intrauterine programming of obesity and type 2 diabetes. Diabetologia. 2019;62(10):1789–18. doi: 10.1007/s00125-019-4951-9
  • Strain WD, Paldánius PM. Diabetes, cardiovascular disease and the microcirculation. Cardiovasc Diabetol. 2018;17(1):57. doi: 10.1186/s12933-018-0703-2
  • Liu Z, Fu C, Wang W, et al. Prevalence of chronic complications of type 2 diabetes mellitus in outpatients - a cross-sectional hospital based survey in urban China. Health Qual Life Outcomes. 2010;8(1):62. doi: 10.1186/1477-7525-8-62
  • Seferović PM, Paulus WJ. Clinical diabetic cardiomyopathy: a two-faced disease with restrictive and dilated phenotypes. Eur Heart J. 2015;36(27):1718–1727. doi: 10.1093/eurheartj/ehv134
  • Kannel WB, Hjortland M, Castelli WP. Role of diabetes in congestive heart failure: the Framingham study. Am J Cardiol. 1974;34(1):29–34. doi: 10.1016/0002-9149(74)90089-7
  • Matsue Y, Suzuki M, Nakamura R, et al. Prevalence and prognostic implications of pre-diabetic state in patients with heart failure. Circ J. 2011;75(12):2833–2839. doi: 10.1253/circj.CJ-11-0754
  • Gustafsson I, Brendorp B, Seibæk M, et al. Influence of diabetes and diabetes-gender interaction on the risk of death in patients hospitalized with congestive heart failure. J Am Coll Cardiol. 2004;43(5):771–777. doi: 10.1016/j.jacc.2003.11.024
  • Miki T, Yuda S, Kouzu H, et al. Diabetic cardiomyopathy: pathophysiology and clinical features. Heart Fail Rev. 2013;18(2):149–166. doi: 10.1007/s10741-012-9313-3
  • Nichols GA, Gullion CM, Koro CE, et al. The incidence of congestive heart failure in type 2 diabetes: an update. Diabetes Care. 2004;27(8):1879–1884. doi: 10.2337/diacare.27.8.1879
  • Rubler S, Dlugash J, Yuceoglu YZ, et al. New type of cardiomyopathy associated with diabetic glomerulosclerosis. Am J Cardiol. 1972;30(6):595–602. doi: 10.1016/0002-9149(72)90595-4
  • Jia G, Hill MA, Sowers JR. Diabetic cardiomyopathy: an update of mechanisms contributing to this clinical entity. Circ Res. 2018;122(4):624–638. doi: 10.1161/CIRCRESAHA.117.311586
  • Nakamura M, Sadoshima J. Cardiomyopathy in obesity, insulin resistance and diabetes. Journal Of Physiology. 2020;598(14):2977–2993. doi: 10.1113/JP276747
  • Penpargkul S, Fein F, Sonnenblick, EH, et al. Depressed cardiac sarcoplasmic reticular function from diabetic rats. J Mol Cell Cardiol. 1981;13(3):303–309. doi: 10.1016/0022-2828(81)90318-7
  • Trost SU, Belke DD, Bluhm WF, et al. Overexpression of the sarcoplasmic reticulum Ca(2+)-ATPase improves myocardial contractility in diabetic cardiomyopathy. Diabetes. 2002;51(4):1166–1171. doi: 10.2337/diabetes.51.4.1166
  • Westermeier F, Riquelme JA, Pavez M, et al. New molecular insights of insulin in diabetic cardiomyopathy. Front Physiol. 2016;7:125. doi: 10.3389/fphys.2016.00125
  • Ritchie RH, Abel ED. Basic mechanisms of diabetic heart disease. Circ Res. 2020;126(11):1501–1525. doi: 10.1161/CIRCRESAHA.120.315913
  • Cai L, Kang YJ. Cell death and diabetic cardiomyopathy. Cardiovasc Toxicol. 2003;3(3):219–228. doi: 10.1385/CT:3:3:219
  • Jia G, DeMarco VG, Sowers JR. Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy. Nat Rev Endocrinol. 2016;12(3):144–153. doi: 10.1038/nrendo.2015.216
  • Boyer JK, Thanigaraj S, Schechtman KB, et al. Prevalence of ventricular diastolic dysfunction in asymptomatic, normotensive patients with diabetes mellitus. Am J Cardiol. 2004;93(7):870–875. doi: 10.1016/j.amjcard.2003.12.026
  • Zabalgoitia M, Ismaeil MF, Anderson L, et al. Prevalence of diastolic dysfunction in normotensive, asymptomatic patients with well-controlled type 2 diabetes mellitus. Am J Cardiol. 2001;87(3):320–323. doi: 10.1016/S0002-9149(00)01366-7
  • Cherney DZ, Odutayo A, Aronson R, et al. Sodium glucose cotransporter-2 inhibition and cardiorenal protection: JACC review Topic of the Week. J Am Coll Cardiol. 2019;74(20):2511–2524. doi: 10.1016/j.jacc.2019.09.022
  • Joubert M, Jagu B, Montaigne D, et al. The Sodium–Glucose Cotransporter 2 Inhibitor Dapagliflozin Prevents Cardiomyopathy in a Diabetic Lipodystrophic Mouse Model. Diabetes. 2017;66(4):1030–1040. doi: 10.2337/db16-0733
  • Qin S-L, Liu S-L, Wang R-R. [Protective effect of capsaicin on against myocardial ischemia-reperfusion injury of rat in vivo]. Sichuan Da Xue Xue Bao. Yi Xue Ban = Journal of Sichuan University. Med Sci Ed. 2008;39(4):550–554.
  • Szallasi A, Cortright DN, Blum CA, et al. The vanilloid receptor TRPV1: 10 years from channel cloning to antagonist proof-of-concept. Nat Rev Drug Discov. 2007;6(5):357–372. doi: 10.1038/nrd2280
  • Tominaga M, Tominaga T. Structure and function of TRPV1. Pflugers Archiv. European J Physiology. 2005;451(1):143–150. doi: 10.1007/s00424-005-1457-8
  • Premkumar LS, Abooj M. TRP channels and analgesia. Life Sci. 2013;92(8–9):415–424. doi: 10.1016/j.lfs.2012.08.010
  • Li L, Chen C, Chiang C, et al. The impact of TRPV1 on cancer pathogenesis and therapy: a systematic review. Int J Biol Sci. 2021;17(8):2034–2049. doi: 10.7150/ijbs.59918
  • Zhang C, Ye L, Zhang Q, et al. The role of TRPV1 channels in atherosclerosis. channels (Austin, Tex. Channels. 2020;14(1):141–150. doi: 10.1080/19336950.2020.1747803
  • Hurt CM, Lu Y, Stary M, et al. Transient Receptor Potential Vanilloid 1 Regulates Mitochondrial Membrane Potential and Myocardial Reperfusion Injury. J Am Heart Assoc. 2016;5(9). doi: 10.1161/JAHA.116.003774
  • Yoshie K, Rajendran, PS, Massoud L, et al. Cardiac TRPV1 afferent signaling promotes arrhythmogenic ventricular remodeling after myocardial infarction. JCI Insight. 2020;5(3). doi: 10.1172/jci.insight.124477
  • Zahner MR, Li D-P, Chen S-R, et al. Cardiac vanilloid receptor 1-expressing afferent nerves and their role in the cardiogenic sympathetic reflex in rats. Journal Of Physiology. 2003;551(2):515–523. doi: 10.1113/jphysiol.2003.048207
  • Hoebart C, Rojas‐Galvan, NS, Ciotu, CI, et al. No functional TRPA1 in cardiomyocytes. Acta Physiol (Oxf). 2021;232(4):e13659. doi: 10.1111/apha.13659
  • Hong J, Lisco AM, Rudebush TL, et al. Identification of Cardiac Expression Pattern of Transient Receptor Potential Vanilloid Type 1 (TRPV1) Receptor using a Transgenic Reporter Mouse Model. Neurosci lett. 2020;737:135320. doi: 10.1016/j.neulet.2020.135320
  • Andrei SR, Sinharoy P, Bratz IN, et al. TRPA1 is functionally co-expressed with TRPV1 in cardiac muscle: co-localization at z-discs, costameres and intercalated discs. channels (Austin, Tex. Channels. 2016;10(5):395–409. doi: 10.1080/19336950.2016.1185579
  • Bizino MB, Jazet IM, Westenberg JJM, et al. Effect of liraglutide on cardiac function in patients with type 2 diabetes mellitus: randomized placebo-controlled trial. Cardiovasc Diabetol. 2019;18(1):55. doi: 10.1186/s12933-019-0857-6
  • Ching L-C, Chen C-Y, Su K-H, et al. Implication of AMP-activated protein kinase in transient receptor potential vanilloid type 1-mediated activation of endothelial nitric oxide synthase. molecular medicine (Cambridge, Mass. 2012;18(5):805–815. doi: 10.2119/molmed.2011.00461
  • Wang Y, Cui L, Xu H, et al. TRPV1 agonism inhibits endothelial cell inflammation via activation of eNOS/NO pathway. Atherosclerosis. 2017;260:13–19. doi: 10.1016/j.atherosclerosis.2017.03.016
  • Ma L, Zhong J, Zhao Z, et al. Activation of TRPV1 reduces vascular lipid accumulation and attenuates atherosclerosis. Cardiovasc Res. 2011;92(3):504–513. doi: 10.1093/cvr/cvr245
  • Luo D, Li W, Xie C, et al. Capsaicin attenuates arterial calcification through promoting SIRT6-mediated deacetylation and degradation of Hif1α (hypoxic-inducible factor-1 alpha). Hypertension. 2022;79(5):906–917. Hypertension (Dallas, Tex. : 1979). doi: 10.1161/HYPERTENSIONAHA.121.18778
  • Parpaite T, Cardouat G, Mauroux M, et al. Effect of hypoxia on TRPV1 and TRPV4 channels in rat pulmonary arterial smooth muscle cells. Pflugers Archiv. Pflugers Arch - Eur J Physiol. 2016;468(1):111–130. doi: 10.1007/s00424-015-1704-6
  • Sun Z, Han J, Zhao W, et al. TRPV1 activation exacerbates hypoxia/reoxygenation-induced apoptosis in H9C2 cells via calcium overload and mitochondrial dysfunction. Int J Mol Sci. 2014;15(10):18362–18380. doi: 10.3390/ijms151018362
  • Gao F, Liang Y, Wang X, et al. TRPV1 activation attenuates high-salt diet-induced cardiac hypertrophy and fibrosis through PPAR-δ upregulation. PPAR Res. 2014;2014:1–12. doi: 10.1155/2014/491963
  • Lang H, Li Q, Yu H, et al. Activation of TRPV1 attenuates high salt-induced cardiac hypertrophy through improvement of mitochondrial function. Br J Pharmacol. 2015;172(23):5548–5558. doi: 10.1111/bph.12987
  • Horton JS, Shiraishi T, Alfulaij N, et al. TRPV1 is a component of the atrial natriuretic signaling complex, and using orally delivered antagonists, presents a valid therapeutic target in the longitudinal reversal and treatment of cardiac hypertrophy and heart failure. Channels (Austin, Tex). 2019;13(1):1–16. doi: 10.1080/19336950.2018.1547611
  • Wei J, Lin J, Zhang J, et al. TRPV1 activation mitigates hypoxic injury in mouse cardiomyocytes by inducing autophagy through the AMPK signaling pathway. Am J Physiol Cell Physiol. 2020;318(5):C1018–C1029. doi: 10.1152/ajpcell.00161.2019
  • Zhong B, Rubinstein J, Ma S, et al. Genetic ablation of TRPV1 exacerbates pressure overload-induced cardiac hypertrophy. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie. 2018;99:261–270. doi: 10.1016/j.biopha.2018.01.065
  • Scotland RS, Chauhan S, Davis C, et al. Vanilloid receptor TRPV1, sensory C-fibers, and vascular autoregulation: a novel mechanism involved in myogenic constriction. Circ Res. 2004;95(10):1027–1034. doi: 10.1161/01.RES.0000148633.93110.24
  • Hoover DB. Effects of capsaicin on release of substance P-like immunoreactivity and physiological parameters in isolated perfused guinea-pig heart. Eur J Pharmacol. 1987;141(3):489–492. doi: 10.1016/0014-2999(87)90571-1
  • Manzini S, Perretti F, de Benedetti L, et al. A comparison of bradykinin- and capsaicin-induced myocardial and coronary effects in isolated perfused heart of guinea-pig: involvement of substance P and calcitonin gene-related peptide release. Br J Pharmacol. 1989;97(2):303–312. doi: 10.1111/j.1476-5381.1989.tb11955.x
  • Szabados T, Gömöri K, Pálvölgyi L, et al. Capsaicin-Sensitive sensory nerves and the TRPV1 ion channel in cardiac Physiology and pathologies. Int J Mol Sci. 2020;21(12):4472. doi: 10.3390/ijms21124472
  • Li J, Zhao H, Supowit SC, et al. Activation of the renin–angiotensin system in α-calcitonin gene-related peptide/calcitonin gene knockout mice. J Hypertens. 2004;22(7):1345–1349. doi: 10.1097/01.hjh.0000125409.50839.f1
  • Wang L, Wang DH. TRPV1 gene knockout impairs postischemic recovery in isolated perfused heart in mice. Circulation. 2005;112(23):3617–3623. doi: 10.1161/CIRCULATIONAHA.105.556274
  • Xiong S, Wang P, Ma L, et al. Ameliorating endothelial mitochondrial dysfunction restores coronary function via Transient receptor potential vanilloid 1–mediated protein kinase A/Uncoupling protein 2 pathway. Hypertension (Dallas, Tex. : 1979). 2016;67(2):451–460. doi: 10.1161/HYPERTENSIONAHA.115.06223
  • McCarty MF, DiNicolantonio JJ, O’Keefe JH. Capsaicin may have important potential for promoting vascular and metabolic health. Open Heart. 2015;2(1):e000262. doi: 10.1136/openhrt-2015-000262
  • Doroshow JH, Locker GY, Myers CE. Enzymatic defenses of the mouse heart against reactive oxygen metabolites: alterations produced by doxorubicin. J Clin Investig. 1980;65(1):128–135. doi: 10.1172/JCI109642
  • Chen Y, Saari JT, Kang YJ. Weak antioxidant defenses make the heart a target for damage in copper-deficient rats. Free Radic Biol Med. 1994;17(6):529–536. doi: 10.1016/0891-5849(94)90092-2
  • Huynh K, Bernardo BC, McMullen JR, et al. Diabetic cardiomyopathy: mechanisms and new treatment strategies targeting antioxidant signaling pathways. Pharmacol Ther. 2014;142(3):375–415. doi: 10.1016/j.pharmthera.2014.01.003
  • Faria A, Persaud SJ. Cardiac oxidative stress in diabetes: mechanisms and therapeutic potential. Pharmacol Ther. 2017;172:50–62. doi: 10.1016/j.pharmthera.2016.11.013
  • Hamblin M, Friedman DB, Hill S, et al. Alterations in the diabetic myocardial proteome coupled with increased myocardial oxidative stress underlies diabetic cardiomyopathy. J Mol Cell Cardiol. 2007;42(4):884–895. doi: 10.1016/j.yjmcc.2006.12.018
  • Di Filippo C, Marfella R, Cuzzocrea S, et al. Hyperglycemia in streptozotocin-induced diabetic rat increases infarct size associated with low levels of myocardial HO-1 during ischemia/reperfusion. Diabetes. 2005;54(3):803–810. doi: 10.2337/diabetes.54.3.803
  • D’Amario D, Migliaro S, Borovac JA, et al. Microvascular dysfunction in heart failure with preserved ejection fraction. Front Physiol. 2019;10:1347. doi: 10.3389/fphys.2019.01347
  • Gambardella J, Sorriento D, Ciccarelli M, et al. Functional role of mitochondria in Arrhythmogenesis. Adv Exp Med Biol. 2017;982:191–202.
  • Aboumsallem JP, Muthuramu I, Mishra M, et al. Effective treatment of diabetic cardiomyopathy and heart failure with reconstituted HDL (Milano) in mice. Int J Mol Sci. 2019;20(6):1273. doi: 10.3390/ijms20061273
  • Moukdar F, Robidoux J, Lyght O, et al. Reduced antioxidant capacity and diet-induced atherosclerosis in uncoupling protein-2-deficient mice. J Lipid Res. 2009;50(1):59–70. doi: 10.1194/jlr.M800273-JLR200
  • Tian XY, Wong WT, Xu A, et al. Uncoupling protein-2 protects endothelial function in diet-induced obese mice. Circ Res. 2012;110(9):1211–1216. doi: 10.1161/CIRCRESAHA.111.262170
  • Dhamrait SS, et al. ?Cardiovascular risk in healthy men and markers of oxidative stress in diabetic men are associated with common variation in the gene for uncoupling protein 2. Eur Heart J. 2004;25(6):468–475. doi: 10.1016/j.ehj.2004.01.007
  • Li L, Chen J, Ni Y, et al. TRPV1 activation prevents nonalcoholic fatty liver through UCP2 upregulation in mice. Pflugers Archiv. European J Physiology. 2012;463(5):727–732. doi: 10.1007/s00424-012-1078-y
  • Lee M-S, Kim, CT, Kim, IH, et al. Effects of capsaicin on lipid catabolism in 3T3-L1 adipocytes. Phytother Res. 2011;25(6):935–939. doi: 10.1002/ptr.3339
  • Sun J, Pu Y, Wang P, et al. TRPV1-mediated UCP2 upregulation ameliorates hyperglycemia-induced endothelial dysfunction. Cardiovasc Diabetol. 2013;12(1):69. doi: 10.1186/1475-2840-12-69
  • Chen J, Zhang Z, Cai L. Diabetic cardiomyopathy and its prevention by nrf2: current status. Diabetes & Metabolism Journal. 2014;38(5):337–345. doi: 10.4093/dmj.2014.38.5.337
  • He X, Kan H, Cai L, et al. Nrf2 is critical in defense against high glucose-induced oxidative damage in cardiomyocytes. J Mol Cell Cardiol. 2009;46(1):47–58. doi: 10.1016/j.yjmcc.2008.10.007
  • Tan Y, Ichikawa T, Li J, et al. Diabetic downregulation of Nrf2 activity via ERK contributes to oxidative stress–induced insulin resistance in cardiac cells in vitro and in vivo. Diabetes. 2011;60(2):625–633. doi: 10.2337/db10-1164
  • Wang Y, Sun W, Du B, et al. Therapeutic effect of MG-132 on diabetic cardiomyopathy is associated with its suppression of proteasomal activities: roles of Nrf2 and NF-κB. Am J Physiol Heart Circ Physiol. 2013;304(4):H567–H578. doi: 10.1152/ajpheart.00650.2012
  • Velmurugan GV, Sundaresan NR, Gupta MP, et al. Defective Nrf2-dependent redox signalling contributes to microvascular dysfunction in type 2 diabetes. Cardiovasc Res. 2013;100(1):143–150. doi: 10.1093/cvr/cvt125
  • Xu Z, Wei Y, Gong J, et al. NRF2 plays a protective role in diabetic retinopathy in mice. Diabetologia. 2014;57(1):204–213. doi: 10.1007/s00125-013-3093-8
  • Lv Z, Xu X, Sun Z, et al. TRPV1 alleviates osteoarthritis by inhibiting M1 macrophage polarization via Ca2+/CaMKII/Nrf2 signaling pathway. Cell Death Dis. 2021;12(6):504. doi: 10.1038/s41419-021-03792-8
  • Flammer AJ, Anderson T, Celermajer DS, et al. The assessment of endothelial function: from research into clinical practice. Circulation. Circulation. 2012;126(6):753–767. doi: 10.1161/CIRCULATIONAHA.112.093245
  • Kitta Y, Obata J-E, Nakamura T, et al. Persistent impairment of endothelial vasomotor function has a negative impact on outcome in patients with coronary artery disease. J Am Coll Cardiol. 2009;53(4):323–330. doi: 10.1016/j.jacc.2008.08.074
  • Moraes RDA, Webb RC, Silva DF. Vascular dysfunction in diabetes and obesity: focus on TRP channels. Front Physiol. 2021;12:645109. doi: 10.3389/fphys.2021.645109
  • Li X, Hou J, Du J, et al. Potential Protective Mechanism in the Cardiac Microvascular Injury. Hypertension. 2018;72(1):116–127. Hypertension (Dallas, Tex. : 1979). doi: 10.1161/HYPERTENSIONAHA.118.11035
  • Zhou Y, Wang X, Guo L, et al. TRPV1 activation inhibits phenotypic switching and oxidative stress in vascular smooth muscle cells by upregulating PPARα. Biochem Biophys Res Commun. 2021;545:157–163. doi: 10.1016/j.bbrc.2021.01.072
  • Zhu S-L, Wang M-L, He Y-T, et al. Capsaicin ameliorates intermittent high glucose-mediated endothelial senescence via the TRPV1/SIRT1 pathway. Phytomedicine. 2022;100:154081. doi: 10.1016/j.phymed.2022.154081
  • Frieler RA, Mortensen RM. Immune cell and other noncardiomyocyte regulation of cardiac hypertrophy and remodeling. Circulation. 2015;131(11):1019–1030. doi: 10.1161/CIRCULATIONAHA.114.008788
  • Mann DL. Innate immunity and the failing heart: the cytokine hypothesis revisited. Circ Res. 2015;116(7):1254–1268. doi: 10.1161/CIRCRESAHA.116.302317
  • Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ Res. 2016;119(1):91–112. doi: 10.1161/CIRCRESAHA.116.303577
  • Yang J, Park Y, Zhang H, et al. Feed-forward signaling of TNF-α and NF-κB via IKK-β pathway contributes to insulin resistance and coronary arteriolar dysfunction in type 2 diabetic mice. Am J Physiol Heart Circ Physiol. 2009;296(6):H1850–H1858. doi: 10.1152/ajpheart.01199.2008
  • Mann DL, Kirschenbaum L. The emerging role of innate immunity in the heart and vascular system: for whom the cell tolls. Circ Res. 2011;108(9):1133–1145. doi: 10.1161/CIRCRESAHA.110.226936
  • Sciarretta S, Paneni F, Palano F, et al. Role of the renin–angiotensin–aldosterone system and inflammatory processes in the development and progression of diastolic dysfunction. Clinical Science (London, England). 2009;116(6):467–477. doi: 10.1042/CS20080390
  • Yan SF, Ramasamy R, Naka Y, et al. Glycation, inflammation, and RAGE: a scaffold for the macrovascular complications of diabetes and beyond. Circ Res. 2003;93(12):1159–1169. doi: 10.1161/01.RES.0000103862.26506.3D
  • Westermann D, Walther T, Savvatis K, et al. Gene deletion of the kinin receptor B1 attenuates cardiac inflammation and fibrosis during the development of experimental diabetic cardiomyopathy. Diabetes. 2009;58(6):1373–1381. doi: 10.2337/db08-0329
  • Fernandes ES, Liang L, Smillie S-J, et al. TRPV1 deletion enhances local inflammation and accelerates the onset of systemic inflammatory response syndrome. Journal Of Immunology (Baltimore, Md). 2012;188(11):5741–5751. doi: 10.4049/jimmunol.1102147
  • Ching L-C, Kou YR, Shyue S-K, et al. Molecular mechanisms of activation of endothelial nitric oxide synthase mediated by transient receptor potential vanilloid type 1. Cardiovasc Res. 2011;91(3):492–501. doi: 10.1093/cvr/cvr104
  • Singh S, Natarajan K, Aggarwal BB. Capsaicin (8-methyl-N-vanillyl-6-nonenamide) is a potent inhibitor of nuclear transcription factor-kappa B activation by diverse agents. J Immun (Baltimore. 1996;157(10):4412–4420. Md. : 1950). doi: 10.4049/jimmunol.157.10.4412
  • Sancho R, Macho A, de La Vega L, et al. Immunosuppressive activity of endovanilloids: N -arachidonoyl-dopamine inhibits activation of the NF-κB, NFAT, and activator protein 1 signaling pathways. Journal Of Immunology (Baltimore, Md). 2004;172(4):2341–2351. doi: 10.4049/jimmunol.172.4.2341
  • Huang W, Rubinstein J, Prieto AR, et al. Transient receptor potential vanilloid gene deletion exacerbates inflammation and atypical cardiac remodeling after myocardial infarction. Hypertension. 2009;53(2):243–250. Hypertension (Dallas, Tex. : 1979). doi: 10.1161/HYPERTENSIONAHA.108.118349
  • Jia G, Habibi J, DeMarco VG, et al. Endothelial mineralocorticoid receptor deletion prevents diet-induced cardiac diastolic dysfunction in females. Hypertension. 2015;66(6):1159–1167. Hypertension (Dallas, Tex.: 1979). doi: 10.1161/HYPERTENSIONAHA.115.06015
  • Ururahy MAG, Loureiro MB, Freire-Neto FP, et al. Increased TLR2 expression in patients with type 1 diabetes: evidenced risk of microalbuminuria. Pediatr Diabetes. 2012;13(2):147–154. doi: 10.1111/j.1399-5448.2011.00794.x
  • Lv Z, Xu X, Sun Z, et al. TRPV1 alleviates osteoarthritis by inhibiting M1 macrophage polarization via Ca/CaMKII/Nrf2 signaling pathway. Cell Death Dis. 2021;12(6):504. doi: 10.1038/s41419-021-03792-8
  • Zhao J-F, Ching L-C, Kou YR, et al. Activation of TRPV1 prevents OxLDL-induced lipid accumulation and TNF-α-induced inflammation in macrophages: role of liver X receptor α. Mediators Inflamm. 2013;2013:1–14. doi: 10.1155/2013/925171
  • Anderson EJ, Kypson AP, Rodriguez E, et al. Substrate-specific derangements in mitochondrial metabolism and redox balance in the atrium of the type 2 diabetic human heart. J Am Coll Cardiol. 2009;54(20):1891–1898. doi: 10.1016/j.jacc.2009.07.031
  • Anderson EJ, Rodriguez E, Anderson CA, et al. Increased propensity for cell death in diabetic human heart is mediated by mitochondrial-dependent pathways. Am J Physiol Heart Circ Physiol. 2011;300(1):H118–H124. doi: 10.1152/ajpheart.00932.2010
  • Kim J-A, Wei Y, Sowers JR. Role of mitochondrial dysfunction in insulin resistance. Circ Res. 2008;102(4):401–414. doi: 10.1161/CIRCRESAHA.107.165472
  • Luo Z, Ma L, Zhao Z, et al. TRPV1 activation improves exercise endurance and energy metabolism through PGC-1α upregulation in mice. Cell Res. 2012;22(3):551–564. doi: 10.1038/cr.2011.205
  • Wang Y-X, Zhang C-L, Yu RT, et al. Regulation of muscle fiber type and running endurance by PPARδ. PLoS Biol. 2004;2(10):e294. doi: 10.1371/journal.pbio.0020294
  • Joyner MJ, Coyle EF. Endurance exercise performance: the physiology of champions. Journal Of Physiology. 2008;586(1):35–44. doi: 10.1113/jphysiol.2007.143834
  • Wu S, Lu Q, Ding Y, et al. Hyperglycemia-Driven inhibition of AMP-Activated protein kinase α2 induces diabetic cardiomyopathy by promoting mitochondria-associated endoplasmic reticulum membranes in vivo. Circulation. 2019;139(16):1913–1936. doi: 10.1161/CIRCULATIONAHA.118.033552
  • Wu S, Lu Q, Wang Q, et al. Binding of FUN14 domain containing 1 with inositol 1,4,5-trisphosphate receptor in mitochondria-associated endoplasmic reticulum Membranes Maintains mitochondrial Dynamics and function in hearts in vivo. Circulation. 2017;136(23):2248–2266. doi: 10.1161/CIRCULATIONAHA.117.030235
  • Wei X, Wei X, Lu Z, et al. Activation of TRPV1 channel antagonizes diabetic nephropathy through inhibiting endoplasmic reticulum-mitochondria contact in podocytes. Metabolism. 2020;105:154182. doi: 10.1016/j.metabol.2020.154182
  • Shimizu M, Umeda K, Sugihara N, et al. Collagen remodelling in myocardia of patients with diabetes. J Clin Pathol. 1993;46(1):32–36. doi: 10.1136/jcp.46.1.32
  • Abed HS, Samuel CS, Lau DH, et al. Obesity results in progressive atrial structural and electrical remodeling: implications for atrial fibrillation. Heart Rhythm. 2013;10(1):90–100. doi: 10.1016/j.hrthm.2012.08.043
  • Gao Y, Kang L, Li C, et al. Resveratrol ameliorates diabetes-induced cardiac dysfunction through AT1R-ERK/p38 MAPK signaling pathway. Cardiovasc Toxicol. 2016;16(2):130–137. doi: 10.1007/s12012-015-9321-3
  • Suematsu Y, Miura S-I, Goto M, et al. LCZ696 , an angiotensin receptor–neprilysin inhibitor, improves cardiac function with the attenuation of fibrosis in heart failure with reduced ejection fraction in streptozotocin-induced diabetic mice. European J Heart Fail. 2016;18(4):386–393. doi: 10.1002/ejhf.474
  • Frangogiannis NG. The role of transforming growth factor (TGF)-β in the infarcted myocardium. J Thoracic Dis. 2017;9(Suppl 1):S52–S63. doi: 10.21037/jtd.2016.11.19
  • Travers JG, Kamal FA, Robbins J, et al. Cardiac fibrosis: the fibroblast awakens. Circ Res. 2016;118(6):1021–1040. doi: 10.1161/CIRCRESAHA.115.306565
  • Wang Q, Zhang Y, Li D, et al. Transgenic overexpression of transient receptor potential vanilloid subtype 1 attenuates isoproterenol-induced myocardial fibrosis in mice. Int J Mol Med. 2016;38(2):601–609. doi: 10.3892/ijmm.2016.2648
  • An D, Rodrigues B. Role of changes in cardiac metabolism in development of diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol. 2006;291(4):H1489–H1506. doi: 10.1152/ajpheart.00278.2006
  • Rijzewijk LJ, van der Meer RW, Lamb HJ, et al. Altered myocardial substrate metabolism and decreased diastolic function in nonischemic human diabetic cardiomyopathy: studies with cardiac positron emission tomography and magnetic resonance imaging. J Am Coll Cardiol. 2009;54(16):1524–1532. doi: 10.1016/j.jacc.2009.04.074
  • Ghosh S, An D, Pulinilkunnil T, et al. Role of dietary fatty acids and acute hyperglycemia in modulating cardiac cell death. Calif.) (Burbank, Los Angeles County. Nutrition. 2004;20(10):916–923. doi: 10.1016/j.nut.2004.06.013
  • Buchanan J, Mazumder PK, Hu P, et al. Reduced cardiac efficiency and altered substrate metabolism precedes the onset of hyperglycemia and contractile dysfunction in two mouse models of insulin resistance and obesity. Endocrinology. 2005;146(12):5341–5349. doi: 10.1210/en.2005-0938
  • Aasum E, Hafstad AD, Severson DL, et al. Age-dependent changes in metabolism, contractile function, and ischemic sensitivity in hearts from db/db mice. Diabetes. 2003;52(2):434–441. doi: 10.2337/diabetes.52.2.434
  • Panchal SK, Bliss E, Brown L. Capsaicin in Metabolic Syndrome. Nutrients. 2018;10(5):630. doi: 10.3390/nu10050630
  • Akiba Y, Kato S, Katsube K-I, et al. Transient receptor potential vanilloid subfamily 1 expressed in pancreatic islet β cells modulates insulin secretion in rats. Biochem Biophys Res Commun. 2004;321(1):219–225. doi: 10.1016/j.bbrc.2004.06.149
  • Tsui H, Razavi R, Chan Y, et al. ‘Sensing’ autoimmunity in type 1 diabetes. Trends Mol Med. 2007;13(10):405–413. doi: 10.1016/j.molmed.2007.07.006
  • Razavi R, Chan Y, Afifiyan FN, et al. TRPV1+ Sensory Neurons Control β Cell Stress and Islet Inflammation in Autoimmune Diabetes. Cell. 2006;127(6):1123–1135. doi: 10.1016/j.cell.2006.10.038
  • Zhong B, Ma S, Wang DH. TRPV1 Mediates Glucose-induced Insulin Secretion Through Releasing Neuropeptides. Vivo (Athens, Greece). 2019;33(5):1431–1437. doi: 10.21873/invivo.11621
  • Wang P, Yan Z, Zhong J, et al. Transient receptor potential vanilloid 1 activation enhances gut glucagon-like peptide-1 secretion and improves glucose homeostasis. Diabetes. 2012;61(8):2155–2165. doi: 10.2337/db11-1503
  • Parlevliet ET, de Leeuw van Weenen JE, Romijn JA, et al. GLP-1 treatment reduces endogenous insulin resistance via activation of central GLP-1 receptors in mice fed a high-fat diet. Am J Physiol Endocrinol Metab. 2010;299(2):E318–E324. doi: 10.1152/ajpendo.00191.2010
  • Borghetti G, von Lewinski D, Eaton DM, et al. Diabetic cardiomyopathy: Current and future therapies. Beyond glycemic control. Front Physiol. 2018;9:1514. doi: 10.3389/fphys.2018.01514
  • Stratton IM, Wei X, Lu Z, et al. Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (UKPDS 35): prospective observational study. BMJ. 2000;321(7258):405–412. doi: 10.1136/bmj.321.7258.405
  • ElSayed NA, Aleppo G, Aroda VR, et al. 9. Pharmacologic approaches to glycemic treatment: standards of Care in diabetes—2023. Diabetes Care. 2023;46(Suppl 1):S140–S157. doi: 10.2337/dc23-S009
  • Chen YH, Feng B, Chen ZW. Statins for primary prevention of cardiovascular and cerebrovascular events in diabetic patients without established cardiovascular diseases: a meta-analysis. Experimental And Clinical Endocrinology & Diabetes: Official Journal, German Society Of Endocrinology [And] German Diabetes Association. 2012;120(2):116–120. doi: 10.1055/s-0031-1297968
  • Varzideh F, Kansakar U, Santulli G. SGLT2 inhibitors in cardiovascular medicine. Eur Heart J Cardiovasc Pharmacother. 2021;7(4):e67–e68. doi: 10.1093/ehjcvp/pvab039
  • Ng K-M, Lau Y-M, Dhandhania V, et al. Empagliflozin Ammeliorates High Glucose Induced-Cardiac Dysfuntion in Human iPSC-Derived Cardiomyocytes. Sci Rep. 2018;8(1):14872. doi: 10.1038/s41598-018-33293-2
  • Wang D, Jiang L, Feng B, et al. Protective effects of glucagon-like peptide-1 on cardiac remodeling by inhibiting oxidative stress through mammalian target of rapamycin complex 1/p70 ribosomal protein S6 kinase pathway in diabetes mellitus. J of Diabetes Invest. 2020;11(1):39–51. doi: 10.1111/jdi.13098
  • Liu J, Liu Y, Chen L, et al. Glucagon-like peptide-1 Analog Liraglutide protects against diabetic cardiomyopathy by the inhibition of the endoplasmic reticulum stress pathway. J Diabetes Res. 2013;2013:1–8. doi: 10.1155/2013/630537
  • Arow M, Waldman M, Yadin D, et al. Sodium–glucose cotransporter 2 inhibitor Dapagliflozin attenuates diabetic cardiomyopathy. Cardiovasc Diabetol. 2020;19(1):7. doi: 10.1186/s12933-019-0980-4
  • Steven S, Oelze M, Hanf A, et al. The SGLT2 inhibitor empagliflozin improves the primary diabetic complications in ZDF rats. Redox Biol. 2017;13:370–385. doi: 10.1016/j.redox.2017.06.009
  • Croteau D, Baka T, Young S, et al. SGLT2 inhibitor ertugliflozin decreases elevated intracellular sodium, and improves energetics and contractile function in diabetic cardiomyopathy. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie. 2023;160:114310. doi: 10.1016/j.biopha.2023.114310
  • Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med. 2015;373(22):2117–2128. doi: 10.1056/NEJMoa1504720
  • Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med. 2017;377(7):644–657. doi: 10.1056/NEJMoa1611925
  • Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2019;380(4):347–357. doi: 10.1056/NEJMoa1812389
  • Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2016;375(4):311–322. doi: 10.1056/NEJMoa1603827
  • Lambadiari V, Pavlidis G, Kousathana F, et al. Effects of 6-month treatment with the glucagon like peptide-1 analogue liraglutide on arterial stiffness, left ventricular myocardial deformation and oxidative stress in subjects with newly diagnosed type 2 diabetes. Cardiovasc Diabetol. 2018;17(1):8. doi: 10.1186/s12933-017-0646-z
  • Otto M, Bucher C, Liu W, et al. 12(S)-HETE mediates diabetes-induced endothelial dysfunction by activating intracellular endothelial cell TRPV1. J Clin Investig. 2020;130(9):4999–5010. doi: 10.1172/JCI136621
  • Otto M, Brabenec L, Müller M, et al. Development of heart failure with preserved ejection fraction in type 2 diabetic mice is ameliorated by preserving vascular function. Life Sci. 2021;284:119925. doi: 10.1016/j.lfs.2021.119925
  • Hennessy E, Rakovac Tisdall A, Murphy N, et al. Elevated 12-hydroxyeicosatetraenoic acid (12-HETE) levels in serum of individuals with newly diagnosed type 1 diabetes. Diabet Med. 2017;34(2):292–294. doi: 10.1111/dme.13177
  • Zhang HJ, Sun CH, Kuang HY, et al. 12S-hydroxyeicosatetraenoic acid levels link to coronary artery disease in type 2 diabetic patients. J Endocrinol Invest. 2013;36(6):385–389. doi: 10.3275/8654
  • Antonipillai I, Nadler, JE, Vu, EJ, et al. A 12-lipoxygenase product, 12-hydroxyeicosatetraenoic acid, is increased in diabetics with incipient and early renal disease. J Clin Endocrinol Metab. 1996;81(5):1940–1945. doi: 10.1210/jc.81.5.1940
  • Fernandes ES, Fernandes MA, Keeble JE. The functions of TRPA1 and TRPV1: moving away from sensory nerves. Br J Pharmacol. 2012;166(2):510–521. doi: 10.1111/j.1476-5381.2012.01851.x