Skip to main content

Therapeutic strategies targeting mechanisms of macrophages in diabetic heart disease

Abstract

Diabetic heart disease (DHD) is a serious complication in patients with diabetes. Despite numerous studies on the pathogenic mechanisms and therapeutic targets of DHD, effective means of prevention and treatment are still lacking. The pathogenic mechanisms of DHD include cardiac inflammation, insulin resistance, myocardial fibrosis, and oxidative stress. Macrophages, the primary cells of the human innate immune system, contribute significantly to these pathological processes, playing an important role in human disease and health. Therefore, drugs targeting macrophages hold great promise for the treatment of DHD. In this review, we examine how macrophages contribute to the development of DHD and which drugs could potentially be used to target macrophages in the treatment of DHD.

Introduction

Diabetes mellitus (DM) is a widespread condition with costly treatment. In 2045, DM is projected to affect 783 million individuals worldwide and incur an annual economic expense of 1054 billion USD [1]. Type 2 DM (T2DM) accounts for 90–95% of all diabetes cases [2], and people with diabetes have more than twice the risk of developing heart failure (HF) than do people without diabetes [3, 4]. DM and its complications are a serious threat to human health and have become a global public health problem [5]. The pathologic mechanisms of different diabetic complications have a high degree of commonality at the vascular level.

One of the most common forms of diabetic heart disease (DHD) is diabetic cardiomyopathy (DCM). Although researchers have devoted significant resources toward developing a treatment for DCM, only a handful of possibilities have emerged, such as using sodium-glucose cotransporter 2 inhibitors (SGLT2is) [6], glucagon-like peptide 1 receptor agonists (GLP1RAs) [7], and metformin [8]. Unfortunately, these treatments have not significantly improved patient prognosis. A central pathogenic mechanism of DHD is the inflammatory response, which exacerbates oxidative stress and endoplasmic reticulum stress [9]. Macrophages, which execute the inflammatory response, influence the course of DHD through the secretion of various inflammatory factors [10]. Therefore, a drug therapy targeting macrophages may be a promising new direction for improved DHD treatment.

Type 1 DM (T1DM) and T2DM, the two primary forms of DM, have different pathogenic mechanisms. T1DM is mainly caused by the autoimmune destruction of pancreatic β-cells and occurs in younger patients, whereas T2DM is caused by insulin resistance and relative insulin deficiency and is usually seen in older and overweight patients. Both T1DM and T2DM can induce DHD and impaired cardiac function. They share some clinical presentations (e.g., diastolic dysfunction, systolic dysfunction, HF) and pathogenic mechanisms (e.g., cardiac hypertrophy and fibrosis, oxidative stress, inflammation, metabolic dysfunction) [11]. Patients with T2DM are more likely to experience ventricular diastolic dysfunction compared to those with T1DM [12]. Although macrophages are involved in DHD, their specific roles in T1DM- and T2DM-induced DHD and possible ways of therapeutic modulation have not been well studied. Common clinical pharmacological treatments, such as with SGLT2is and GLP1RAs, have not been studied in cardiovascular outcomes trials in patients with T1DM, despite studies showing that these treatments can lower hemoglobin A1c (HbA1c) levels, reduce body weight, facilitate glucose control, and reduce insulin dose requirements [13,14,15]. In the present review, we discuss the role of macrophages in DHD and their pharmacological targeting, with a focus on T2DM-related DHD.

Overview of macrophages: origin, polarization, and function

Macrophages are evolutionarily conserved innate immune cells that play an essential role in human health and disease [16]. Macrophages are heterogeneous, with phenotypes and functions that are regulated by the surrounding microenvironment. Macrophage polarization is the process by which macrophages show specific phenotypic and functional responses upon microenvironmental stimulation and signaling [17]. M0 macrophages are induced by macrophage colony-stimulating factor (M-CSF) to differentiate from monocytes and represent the initial state of macrophage polarization [18]. Depending on the type of stimulus and the pattern and functional properties of surface molecules and secreted cytokines, M0 macrophages polarize into one of two main subpopulations: M1 and M2 macrophages [19]. An illustration of macrophage polarization is shown in Fig. 1. M1 macrophages have potent anti-microbial and anti-tumor activities and participate in the clearance of pathogens by activating nicotinamide adenine dinucleotide phosphate oxidase (NOX) and producing reactive oxygen species (ROS) during injury or infection [20]. Alternately activated or M2 macrophages, which have anti-inflammatory and immunomodulatory effects, are polarized by T helper 2 (Th2) cytokines interleukin-4 (IL-4) and IL-13 [21,22,23]. M2 macrophages have a strong phagocytic ability, can remove dead cells, promote tissue repair and wound healing, and have pro-angiogenic and profibrotic properties [24].

Fig. 1
figure 1

A schematic representation of macrophage polarization. Monocytes can be differentiated into M0 macrophages in response to M-CSF stimulation. M0 macrophages can be polarized into M1 macrophages in response to LPS or Th1 cytokines (INF-γ, TNF-α). M0 macrophages are polarized into M2 macrophages in response to Th2 cytokines (IL-4, IL-13). Depending on the stimuli, M2 macrophages are subdivided into M2a, M2b, M2c, and M2d macrophages. Abbreviations: M-CSF: macrophage colony-stimulating factor, INF-γ: interferon-γ, TNF-α: tumor necrosis factor-α, LPS: lipopolysaccharide, IL: interleukin, TGF-β: transforming growth factor-β, TLR: toll-like receptor

M1 and M2 macrophages are not two distinct cell populations [25] but are present in a spectrum of different functional states, including pro-inflammatory and anti-inflammatory phenotypes of the cells [26]. The concept of M1/M2 is based on several in vitro studies in which macrophages were stimulated in culture with a defined set of factors [27]. Compared to in vivo situations, macrophages cultured outside their native tissue microenvironment have significant differences in their polarization and function [28]. Given the complexity of M1/M2 polarization [27], the functional and stimulant-based understanding of macrophages in the context of DHD warrants further exploration.

The role of macrophages in diabetic heart disease

According to the currently accepted pathophysiologic mechanisms of DHD [29,30,31,32], insulin resistance and metabolic derangements induce hyperglycemic environments in patients with DM [33]. As the disease progresses, the microenvironment promotes inflammatory responses, oxidative stress, advanced glycation end product (AGE) production, and renin-angiotensin-aldosterone system (RAAS) activation. These pathophysiologic abnormalities promote cardiac stiffness, hypertrophy, and fibrosis, leading to diastolic dysfunction, systolic dysfunction, and HF. Below, we discuss the role of macrophages in these pathophysiologic mechanisms, as illustrated in Fig. 2.

Fig. 2
figure 2

Schematic diagram of the mechanisms involved in the development of DHD by macrophages. The hyperglycemic environment in diabetic patients induces CM injury, and necrotic CMs activate mast cells by releasing DAMPs, which induce the secretion of TNF-α and IL-1β, leading to the activation of ECs. Activated ECs induce monocytes to differentiate into CCR2+ macrophages and CCR2-macrophages; the former create an inflammatory environment and remove necrotic tissue, while the latter release IL-10 and TGF-β1, which activate fibroblasts to differentiate into myofibroblasts and produce ECM and collagen for tissue repair. The hyperglycemic environment also directly activates ECs, which secrete cytokines and chemokines that induce macrophage polarization to the M1 phenotype. In turn, M1 macrophages secrete inflammatory factors that promote cardiac inflammatory responses and insulin resistance. In addition, a hyperglycemic environment leads to increased production of AGEs, and the overproduction of AGEs promotes ROS production. The hyperglycemic environment can also act on HMGB1, gradually activating their downstream reactions, leading to increased ROS generation, and exacerbating oxidative stress. Furthermore, ATMs can secrete miRNA-containing exosomes that act on insulin-target cells to develop insulin resistance. Abbreviations: CM: cardiomyocyte, DAMP: danger-associated molecular pattern, TNF-α: tumor necrosis factor-α, IL: interleukin, EC: endothelial cell, TGF-β1: transforming growth factor-β1, ECM: extracellular matrix, AGE: advanced glycation end product, ROS: reactive oxygen species, HMGB1: high mobility group box 1, ATM: adipose tissue macrophage, NF-κB: nuclear factor kappa-B, NLRP3: NLR family pyrin domain-containing protein 3, TLR4: toll-like receptor 4

Cardiac inflammation

DM is characterized by hyperglycemia and insulin resistance, which promote an inflammatory state in the body [9]. The inflammatory response is a critical mechanism by which the heart responds to injury and undergoes adaptive remodeling [34]. The hyperglycemic environment in patients with DM affects protein glycation and the production of AGEs, which activate the nuclear factor kappa B (NFκB) pathway in macrophages. This activation, in turn, induces the production of inflammatory cytokines, leading to the activation of macrophages toward an inflammatory phenotype [35,36,37]. The dysregulation of macrophages between pro-inflammatory and anti-inflammatory phenotypes promotes excessive inflammation and cardiac injury [38]. In individuals with diabetes, macrophages secrete cytokines and chemokines that play a role in developing cardiomyocyte hypertrophy and extracellular matrix (ECM) remodeling. Stimulated by prolonged hyperglycemia, pro-inflammatory macrophages and lymphocytes accumulate and infiltrate into the heart, promoting the secretion of cytokines, such as tumor necrosis factor (TNF), IL-6, IL-1β, interferon-γ (INF-γ), and transforming growth factor-β (TGF-β), which can induce or exacerbate cardiac injury, leading to further adverse remodeling and myocardial fibrosis [39,40,41,42].

High mobility group box 1 (HMGB1) is a nuclear chromatin protein that interacts with nucleosomes, transcription factors, and histones to promote the transcription of many genes associated with inflammatory processes [43, 44]. More precisely, in a hyperglycemic environment, HMGB1 can activate toll-like receptor 4 (TLR4) [45]. TLR4 is expressed in cardiac inflammatory cells, cardiac fibroblasts, and cardiomyocytes [46], and its role in mediating inflammatory signaling in the development of DHD has been demonstrated in animal models [47,48,49,50].

Insulin resistance

Inflammation can lead to the development of insulin resistance. Studies have shown that inhibiting inflammatory response pathways can greatly enhance insulin sensitivity in both animals and humans [51, 52]. TNF-α and monocyte chemoattractant protein 1 (MCP1) play important roles in this process. TNF-α is involved in insulin resistance by activating inflammatory kinases, including c-Jun N-terminal kinase (JNK) [53] and inhibitor kappa B kinase (IKK) [54]. Previous studies have shown that increased TNF-α and macrophage levels in the adipose tissue (AT) of obese mice and humans are associated with insulin resistance [55,56,57]. Macrophages, especially adipose tissue macrophages (ATMs), which are a significant source of pro-inflammatory cytokines, contribute to reduced insulin sensitivity in a paracrine and potentially endocrine manner [58]. Reduction of ATMs or TNF-α by their depletion or ablation of MCP1 or its receptor in mice was associated with improved insulin sensitivity [59,60,61]. In addition, hepatic insulin resistance is observed with increased TNF-α expression in AT when macrophages are recruited to AT by the overexpression of MCP1. Moreover, knockout of MCP1 protected against high-fat-diet (HFD)-induced insulin resistance [60].

ATMs can also affect insulin sensitivity and glucose homeostasis via non-inflammatory pathways. Obesity triggers lysosome biogenesis, resulting in lipid catabolism and buildup of ATMs, specifically of CD11c+ macrophages [62] or Trem2+ macrophages [63]. Deficiency of Trem2 in mice on HFD reduces lipid accumulation in Trem2+ ATMs. These macrophages contribute to aggravated glucose intolerance [63]. Consistently, lipid storage in ATMs can regulate systemic glucose homeostasis and insulin sensitivity [64].

Myocardial fibrosis

Cardiac fibrosis is an unavoidable consequence of chronic myocardial injury and is characterized by the accumulation of ECM proteins in the cardiac interstitium [65]. Intracellular signaling and crosstalk between resident cardiac macrophages and other cells play a crucial role in the onset, propagation, and progression of cardiac inflammation, tissue remodeling, and myocardial fibrosis [66]. The two main types of resident cardiac macrophages are C-C motif chemokine receptor 2 (CCR2)+ and CCR2- macrophages. CCR2+ macrophages maintain their numbers primarily through monocyte recruitment and are mainly involved in pro-inflammatory responses, whereas CCR2- macrophages are primarily embryonic in origin and play a role in promoting angiogenesis and tissue repair [67]. Early in myocardial injury, cardiomyocytes undergo necrosis and release endogenous danger-associated molecular patterns (DAMPs). DAMPs activate mast cells to release pro-inflammatory cytokines, such as TNF-α and IL-1β, as well as chemokines, which activate endothelial cells and recruit activated CCR2+ monocytes and neutrophils. CCR2+ monocytes differentiate into macrophages at the site of injury and work with neutrophils to create an inflammatory environment and remove necrotic tissue [68]. In contrast to CCR2 + macrophage function, CCR2- macrophages are activated in response to myocardial injury and release IL-10 and TGF-β1 [65], which activate fibroblasts to differentiate into myofibroblasts that produce ECM and collagen for tissue repair. However, cardiomyocyte death and increased cardiac inflammation lead to enhanced activation of myofibroblasts and uncontrolled production of ECM and collagen, which accumulate in damaged cardiac tissue and eventually lead to myocardial fibrosis [69, 70].

In addition, the imbalance of M1 and M2 macrophages may be an important factor in promoting myocardial fibrosis. In the context of DHD, AGEs have the ability to direct macrophages toward a pro-inflammatory M1 phenotype, which leads to myocardial injury and, ultimately, myocardial fibrosis [71].

Oxidative stress

Oxidative stress occurs from an imbalance in the generation of free radicals and antioxidants [72]. The overproduction of ROS is believed to be a central mechanism of cardiac inflammation and remodeling [73, 74] and contributes to oxidative stress in both the early and late stages of DCM [75, 76]. In a hyperglycemic environment, HMGB1 can activate TLR4 [45]. Activation of TLR4 leads to the activation of NFκB and the NLR family pyrin domain-containing protein 3 (NLRP3) inflammasome, which recruits procaspase-1 [77, 78]. Activated caspase-1 serves as an enhancer of multiple pro-inflammatory pathways involving NFκB, chemokines, and ROS. An NFκB positive-feedback loop further increases NLRP3 inflammasome assembly and procaspase-1 activation, which stimulates additional ROS production [78].

Another key enzyme in ROS production is NOX [79]. In the diabetic heart, hyperglycemia leads to aberrant activation of mitochondrial NOX, which exacerbates ROS production [80]. In addition, macrophages under hyperglycemic conditions have reduced glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activity, leading to the increased formation of AGEs and their precursors [81]. AGEs can be released extracellularly and bind to AGE receptors (RAGEs) in an autocrine and paracrine manner, resulting in increased ROS production [82]. The interaction between AGEs and RAGEs also promotes M1 polarization by inducing the secretion of IL-6 and TNF-α, which further exacerbates ROS production [83].

Trained immunity

Both T1DM and T2DM are characterized by hyperglycemia, and the primary focus of treatment is to lower blood glucose levels. However, some studies have found that the risk of cardiovascular complications persists in some diabetic patients even after glucose-lowering therapy [84]. This phenomenon is referred to as a “legacy effect” or “metabolic memory” [85]. Innate myeloid cells, such as macrophages and monocytes, have the potential for enhanced responsiveness to secondary stimulation, a phenomenon referred to as “trained immunity” [86]. Trained immunity is the process by which innate immune cells undergo long-term functional reprogramming after brief exposure to a stimulus, which persists after removing that stimulus [87]. The “legacy effect” may be related to this “trained immunity” of macrophages. Edgar et al. [88] found that bone marrow-derived macrophages (BMDMs) cultured in a high-glucose environment had increased glycolysis and enhanced polarization toward the M1 phenotype. The BMDMs isolated from diabetic mice, when cultured at physiological glucose concentrations, had persistent pro-inflammatory status despite normalization of the external glucose concentration. Therefore, therapies that target macrophage-trained immunity may offer a potential approach to treating DHD.

Pharmacological targeting of macrophages in DHD

DHD includes both coronary artery disease (CAD) and DCM. Therapeutic strategies for CAD are well established, whereas therapeutic measures for DCM, in sharp contrast, are lacking. The plasticity and adaptability of macrophages in response to various stimuli make them attractive targets for pharmacotherapy. Next, we will summarize several drugs that modulate macrophage function and have the potential to treat DHD.

Sodium-glucose cotransporter 2 inhibitors (SGLT2is)

Sodium-glucose cotransporters (SGLTs) promote the reabsorption of blood glucose after glomerular filtration in the proximal tubules of the kidney [89]. Two types of SGLT have been identified: SGLT1 and SGLT2. SGLT2 regulates about 90% of glucose reabsorption, with the remainder accomplished by SGLT1 [90].

SGLT2is selectively block SGLT2 activity on the proximal renal tubule, leading to the removal of excess glucose in the urine and ultimately resulting in lower blood glucose levels. Additionally, SGLT2is reduce glycosylated proteins, improve insulin sensitivity, and enhance beta cell function [91]. SGLT2is were first introduced as medications for managing diabetes. They were also shown to provide benefits, such as reducing body weight and decreasing blood pressure [92]. More critically, treatment with SGLT2is has been shown to reduce the risk of cardiovascular disease in patients with T2DM when compared with placebo treatment [93, 94]. There is much evidence to suggest that SGLT2is may have the potential to reduce inflammatory responses [95,96,97,98], myocardial fibrosis [95], insulin resistance [99, 100], and oxidative stress [101, 102], making them a promising treatment option for DHD. SGLT2is-related clinical studies are shown in Table 1; SGLT2is-related preclinical studies are shown in Table 2.

Table 1 Clinical studies of SGLT2is for the treatment of DM or DM-related diseases
Table 2 Preclinical studies of SGLT2is for the treatment of DM or DM-related diseases

Glucagon-like peptide 1 receptor agonists (GLP1RAs)

Glucagon-like peptide 1 (GLP-1) is produced by the posttranslational proteolytic cleavage of proglucagon protein [110]. GLP-1 can enhance insulin secretion in a glucose-dependent manner by activating the GLP-1 receptor (GLP1R) that is highly expressed on islet β cells [111]. GLP1RAs, the drugs used to treat patients with diabetes, also provide benefits for patients with cardiovascular disease. Obesity and diabetes are both risk factors for cardiovascular disease. According to a study by Lazzaroni et al. [112], GLP1RAs significantly reduced body weight in patients with T2DM. A systematic review and meta-analysis of cardiovascular outcome trials of GLP1RAs showed that GLP1RA treatment reduced major adverse cardiovascular events (MACE) by 12% and reduced all-cause mortality by 12% [113]. Furthermore, in patients treated with GLP1RAs, HF admission rates were reduced by 9% [113]. However, in another meta-analysis, GLP1RAs had no significant effect on HF admission rates [114]. Thus, although the benefit of GLP1RAs for patients with cardiovascular disease has been established, the benefits are less certain for patients with HF. Because advanced DHD is characterized by HF [32, 115], GLP1RAs may have little effect on patients with advanced DHD. However, whether GLP1RAs are effective in treating patients with early DHD or preventing the development of DHD warrants further investigation.

Preclinical studies have shown that GLP1RAs have the potential to treat DHD. The GLP1RA liraglutide increases the myocardial glucose oxidation rate and alleviates DCM in C57BL/6J mice [116]. Furthermore, liraglutide-treated rats with DCM showed reduced inflammation, myocardial fibrosis, and apoptosis [117]. Researchers showed that using the GLP1RA exenatide to treat mice with DCM significantly improved serum B-type natriuretic peptide, myocardial fibrosis, myocardial lipid deposition, and echocardiographic parameters [7]. Other researchers showed that treatment with the novel oral GLP1RA oral hypoglycemic peptide 2 (OHP2) prevents DCM in rats by alleviating cardiac lipotoxicity-induced mitochondrial dysfunction [118].

GLP1RAs treatment of DCM may be closely related to their interactions with macrophages. Under pathological stress, macrophages contribute to an excessive inflammatory response, which leads to insulin resistance and diabetes. GLP1RAs can attenuate macrophage infiltration and inhibit the expression of IL-1β, IL-6, and TNF-α [119]. In addition, GLP1RAs can inhibit macrophage polarization to the M1 pro-inflammatory phenotype [120] and promote macrophage polarization to the M2 anti-inflammatory phenotype [121, 122]. The large amount of pro-inflammatory cytokines and chemokines in adipose tissue is a key factor contributing to insulin resistance in patients with T2DM [60, 123, 124]. GLP1RAs inhibit inflammatory mediators in adipose tissue and contribute to improved insulin sensitivity [125]. Moreover, GLP1RAs demonstrate a direct protective effect on the development of diabetes-associated myocardial fibrosis and diastolic dysfunction [126]. Taken together, GLP1RAs contribute to relieving DHD by modulating macrophage function. GLP1RAs-related clinical studies are shown in Table 3; GLP1RAs-related preclinical studies are shown in Table 4.

Table 3 Clinical studies of GLP1RAs for the treatment of DM or DM-related diseases
Table 4 Preclinical studies of GLP1RAs for the treatment of DM or DM-related diseases

Metformin

Metformin is a first-line medication for treating patients with T2DM [134] and works by decreasing the production of glucose in the liver and activating the adenosine monophosphate-activated protein kinase (AMPK). Because metformin activates AMPK, it may also induce the regression of myocardial hypertrophy [31]. In addition, metformin enhances insulin sensitivity by increasing the activity of insulin receptor tyrosine kinase, thereby promoting glycogen synthesis and improving the recruitment and activity of the glucose transporter 4 (GLUT4) [135]. In a mouse model of transverse aortic constriction (TAC)-induced HF, treatment with metformin attenuated myocardial fibrosis by inhibiting the TGFβ1-Smad3 signaling pathway [136]. In addition, clinical studies have shown that metformin treatment reduced the incidence of HF in patients with diabetes [137, 138]. Collectively, this evidence suggests that metformin has potential for use in the treatment of DHD.

Studies have shown that metformin provides benefits by its effects on macrophages. In human macrophages, treatment with metformin selectively inhibited the differentiation of human monocytes into pro-inflammatory M1 macrophages [139]. In addition, LPS stimulated M2 macrophages to produce ROS that are harmful to surrounding tissues, a process that was inhibited by the addition of metformin [139]. Interestingly, metformin reduces oxidative stress and inflammatory responses by inhibiting the differentiation of human monocytes into M1 macrophages and limiting macrophage ROS production through the activation of AMPK [139]. Metformin can also exert anti-inflammatory effects by modulating the AMPK/mTOR signaling pathway to inhibit activation of the NLRP3 inflammasome and favor macrophage polarization toward the M2 phenotype [140]. These findings suggest that metformin acts as a cardioprotective and anti-inflammatory agent by stimulating AMPK/autophagy and thus inhibiting the NLRP3 inflammasome, which is closely associated with macrophages in DHD [141].

HMGB1 is released by necrotic cells and is a potential target for the development of anti-inflammatory therapies [142]. Metformin was found to significantly reduce the inflammatory response of LPS-stimulated macrophages in mice (in vivo) and in RAW 264.7 cells (in vitro) by inhibiting HMGB1 secretion [143]. Guo et al. [144] showed that metformin alleviated olanzapine-induced insulin resistance by inhibiting macrophage infiltration and polarization-mediated inflammatory responses in white adipose tissue of rat epididymis. Cortés et al. [145] found that the anti-inflammatory and inhibitory effects of metformin on ROS were dependent on the expression of the plasticity factor ZEB1 in macrophages. Therefore, metformin may potentially be used to treat DHD. Metformin-related clinical studies are shown in Table 5; metformin-related preclinical studies are shown in Table 6.

Table 5 Clinical studies of metformin for the treatment of DM or DM-related diseases
Table 6 Preclinical studies of metformin for the treatment of DM or DM-related diseases

Renin-angiotensin-aldosterone system inhibitors (RAASis)

Activation of the RAAS in patients with diabetes leads to inflammation, cardiac fibrosis, and oxidative stress, all of which contribute to cardiac remodeling and can be reversed or prevented by RAAS blockade [152, 153]. RAASis include angiotensin-converting enzyme inhibitors (ACEis), angiotensin II receptor blockers (ARBs), renin inhibitors, and mineralocorticoid receptor antagonists (MRAs) [154]. ACEis reduce cardiovascular disease incidence and all-cause mortality and increase cellular insulin sensitivity in patients with diabetes [155]. In a rat model of ischemic cardiomyopathy, the ARB valsartan attenuates TLR activity, inhibits NFκB activity, and reduces circulating cytokine levels [156]. Candesartan, another ARB, ameliorates abnormal local calcium release from cardiomyocytes in the atrial tissue of rats with DCM [157]. Renin inhibitors, such as aliskiren, improve left ventricular hypertrophy and end-systolic volume in patients with diabetes [158, 159]. These studies suggest that RAASis have the potential to treat DHD. The activation of RAAS contributes to the infiltration of macrophages, and the antagonistic macrophage mineralocorticoid receptors have a significant protective effect on cardiovascular remodeling [160]. Activated macrophages produce angiotensin-converting enzyme (ACE), which induces local expression of angiotensin II (Ang II). The ability of ACEis to reduce left ventricular mass and ameliorate myocardial fibrosis suggests a direct link between macrophages, macrophage-derived ACE, myofibroblasts, and left ventricular remodeling [161]. Studies have shown that aldosterone induces galectin-3 expression in macrophages and vascular endothelial cells that leads to vascular and cardiac fibrosis, implying a correlation between RAAS and macrophages [162]. In addition, adipose-infiltrating macrophages have been shown to secrete pro-inflammatory cytokines, such as IL-6 and TNF-α, which trigger activation of the RAAS [163]. In summary, a positive-feedback mechanism may exist between macrophages and the RAAS, leading to mutual activation; therefore, RAASis may indirectly inhibit macrophage hyperactivation, resulting in a therapeutic effect on DHD. RAASis-related clinical studies are shown in Table 7; RAASis-related preclinical studies are shown in Table 8.

Table 7 Clinical studies of RAASis for the treatment of DM or DM-related diseases
Table 8 Preclinical studies of RAASis for the treatment of DM or DM-related diseases

β2-adrenergic receptor agonists (β2ARAs)

G protein–coupled receptors (GPCRs) are important proteins that mediate most cellular responses to external stimuli [167]. Adrenergic receptors (ARs), belonging to the GPCR family, contain both α and β subtypes. β ARs can be further subdivided into β1, β2, and β3 ARs, which are located on the surface of effector cells. β2AR has been extensively studied and is found predominantly in human smooth muscle, where it regulates a variety of physiologic processes [168, 169]. Many β2ARAs have been developed. Because β2ARAs induce relaxation of airway smooth muscle, they are used to treat various respiratory diseases, particularly asthma and chronic obstructive pulmonary disease (COPD) [170]. However, many potential uses for β2ARAs remain to be explored, such as in the treatment of DHD.

In a screening of 1040 compounds with anti-inflammatory effects in rat bone marrow macrophages, researchers identified β2ARA as the most potent compound in inhibiting NFκB-dependent pro-inflammatory TNF-α production by macrophages [171]. Subsequently, they found that β2ARAs inhibited TNF-α production in peripheral blood mononuclear cells of streptozotocin (STZ)-induced diabetic rats [171]. To elucidate the mechanism, they exposed human monocytic leukemia cells and bone marrow macrophages to a high-glucose environment. High glucose reduced the expression of β-arrestin2, a negative regulator of NFκB activation, and its interaction with IκBα, subsequently enhancing the phosphorylation of IκBα and the activation of NFκB. The addition of β2ARAs enhanced the expression of β-arrestin2 and its interaction with IκBα, which led to the downregulation of NFκB. siRNA specific for β-arrestin2 reversed the β2ARA-mediated inhibition of NFκB activation and inflammatory cytokine production. In addition, Zucker diabetic fatty (ZDF) rats treated with the β2ARA salbutamol for 12 weeks showed attenuated monocyte activation, as well as pro-inflammatory and profibrotic responses in the kidneys and heart [172]. Several other studies [171, 173] have shown beneficial effects of β2ARAs in treating DM and its complications. Notably, inhibiting macrophage activation and cardiomyopathy progression with β2ARAs only occurs under hyperglycemic conditions and not normoglycemic conditions [174]. In summary, β2ARAs may be promising drugs for treating DHD. β2ARAs-related clinical studies are shown in Table 9; β2ARAs-related preclinical studies are shown in Table 10.

Table 9 Clinical studies of β2ARAs for the treatment of DM or DM-related diseases
Table 10 Preclinical studies of β2ARAs in DM or DM-related diseases

Potential therapeutic strategy

Targeting MicroRNAs (MiRNAs)

MicroRNAs (miRNAs) are small RNA molecules that are important regulators of different cellular processes. MiRNAs control gene expression at the post-transcriptional level by disassembling or inhibiting the translation of target messenger RNAs (mRNAs) by binding to their 3’-untranslated region (3’UTR) [178].

MiRNAs play important roles in many aspects of macrophage biology, particularly in immune cells, such as monocytes and macrophages [179]. They regulate polarization, differentiation, inflammation, and phagocytosis [180]. For example, miR-720 and miR-127 promote M1 macrophage polarization and suppress M2 polarization by targeting GATA binding protein 3 (GATA3) and B-cell lymphoma 6 (BCL6), respectively, which are critical for M2 polarization [181, 182]. MiR-155 promotes M1 polarization. Both gain-of-function and loss-of-function studies in vivo have shown that miR-155 is necessary for the typical development of the macrophage inflammatory state [183]. Abdominal macrophages that overexpress miR-146a showed an increase in M2-type marker genes (e.g., cluster of differentiation 206 (CD206), arginase 1 (ARG1), C-C motif chemokine ligand 22 (CCL22), and CCL17) and a decrease in M1-type phenotypic markers (e.g., inducible nitric oxide synthase, IL-12, IL-6, TNF, and CD86) [184], indicating that miR-146a may have anti-inflammatory effects.

MiRNAs have been shown to be involved in the process of developing DCM. Myocyte enhancer factor 2 C (MEF2C) is a key transcription factor in promoting cardiomyocyte hypertrophy [185]. In mice and rats with DCM, both miR-133a and miR-373 are involved in MEF2C signaling, leading to cardiomyocyte hypertrophy and mediating cardiac fibrosis through the activation of the p300 gene [185, 186]. In addition, miR-208a [187] and miR-451 [188] are involved in cardiomyocyte hypertrophy. Liu et al. [189] showed that miR-21 levels were significantly elevated in cardiac fibroblasts treated with high glucose, leading to increased collagen synthesis and elevated phosphorylated p38 mitogen-activated protein kinase (MAPK). In addition, inhibiting miR-21 by blocking the activation of the p38 signaling pathway reduced fibrosis, suggesting that miR-21 plays a critical role in DCM. Moreover, miRNAs are associated with oxidative stress in rats with DCM. The significant downregulation of miR-499, miR-1, and miR-133 was observed in high glucose–treated cardiomyocytes, which was reversed by treatment with the antioxidant N-acetylcysteine, suggesting that the downregulation of these miRNAs in the diabetic heart is caused by oxidative stress [190].

Perhaps more importantly, miRNAs modulate macrophage polarization that occurs during the development of DCM. Wang and colleagues showed that miR-657 regulates inflammation and insulin resistance in patients with diabetes by targeting the FAM46C gene to promote macrophage polarization toward M1 [191]. A study discovered that miR-223, a significant regulator of macrophage polarization, suppresses macrophage activation toward a pro-inflammatory phenotype by inhibiting Pknox1. This prevents high-fat-diet-induced adipose tissue inflammatory response and systemic insulin resistance in mice [192]. Thus, miRNAs may control the inflammatory response and insulin resistance by regulating macrophage polarization. Furthermore, miRNAs may be involved in the pathogenesis of DCM by regulating genes related to cardiomyocyte hypertrophy, oxidative stress, and cardiac fibrosis. Together, these findings suggest that targeting miRNAs may be a novel therapeutic strategy for the treatment of DCM. MicroRNAs-related preclinical studies are shown in Table 11.

Table 11 Preclinical studies of microRNAs in DM or DM-related cardiovascular diseases

Melatonin

Melatonin, an endogenous indoleamine hormone with potential free radical scavenging ability, is synthesized and secreted by the pineal gland in mammals and is primarily involved in physiological activities associated with the light-dark cycle [203]. Because of their wide distribution in the body, melatonin receptors exhibit a variety of biological activities beyond antioxidant activity, such as anti-inflammatory effects [204] and regulation of insulin secretion [205]. Fiorina et al. [206] showed that changes in melatonin secretion are related to the immune status of the body. In addition, melatonin may protect against cardiac complications in patients with DM by attenuating apoptotic pathways and addressing the inflammatory response and ROS burden by promoting macrophage polarization toward an anti-inflammatory state [207]. Further studies on melatonin-targeted regulation of macrophages in DHD should be explored.

Conclusion

DHD is a serious complication of DM, posing a significant global health burden. Decades have passed since the discovery of DHD, but limited progress has been made in elucidating the mechanisms and identifying the therapeutic targets of DHD. Macrophages are crucial immune cells that play an indispensable role in maintaining normal physiological functions and resisting diseases. However, their role in the development of DHD has rarely been studied. In this review, we emphasized the significant role of macrophages in the pathogenesis of DHD. Macrophages play a major role in the development of DHD through mechanisms that primarily cause cardiac inflammation, insulin resistance, myocardial fibrosis, and oxidative stress. Additionally, the body’s hyperglycemic environment induces macrophage-trained immunity, which further contributes to the development of DHD. Finally, we present five classes of drugs that may have therapeutic effects on DHD by modulating macrophage function: SGLT2is, GLP1RAs, metformin, RAASis, and β2ARAs. Additionally, miRNAs may be a novel therapeutic for treating DHD, capable of targeting and modulating macrophages. In summary (Fig. 3), macrophages hold great promise as therapeutic targets for DHD and have opened an exciting avenue for developing novel treatments for DHD.

Fig. 3
figure 3

Summary schematic. Inflammation, myocardial fibrosis, insulin resistance, and oxidative stress are the four core mechanisms leading to DHD, and macrophages may play an essential role in these mechanisms. Five types of drugs—SGLT2is, GLP1RAs, metformin, RAASis, and β2ARAs—may have therapeutic potential by regulating macrophages. MiRNAs, which also regulate macrophages, may be involved in the pathogenesis of DHD and may also be potential targets for the treatment of DHD. In addition, melatonin is a promising research direction for treating DHD. Abbreviations: ACE: angiotensin-converting enzyme, AMPK: adenosine monophosphate-activated protein kinase, BW: body weight, BP: blood pressure, CV: cardiovascular, IL: interleukin, MCP-1: monocyte chemoattractant protein-1, NF-κB: nuclear factor kappa-B, ROS: reactive oxygen species, TLR: toll-like receptor, TNF-α: tumor necrosis factor-α

Data availability

Not applicable.

Abbreviations

ACE:

Angiotensin-converting enzyme

ACEi:

Angiotensin-converting enzyme inhibitor

AGE:

Advanced glycation end product

AMPK:

Adenosine monophosphate-activated protein kinase

Ang:

Angiotensin

AR:

Adrenergic receptor

ARA:

Adrenergic receptor agonist

ARB:

Angiotensin II receptor blocker

ARG:

Arginase

ATM:

Adipose tissue macrophage

BCL:

B-cell lymphoma

BMDM:

Bone marrow-derived macrophage

CAD:

Coronary artery disease

CCR:

C-C motif chemokine receptor

CCL:

C-C motif chemokine ligand

CD:

Cluster of differentiation

COPD:

Chronic obstructive pulmonary disease

DAMP:

Danger-associated molecular pattern

DCM:

Diabetic cardiomyopathy

DHD:

Diabetic heart disease

DM:

Diabetes mellitus

ECM:

Extracellular matrix

GAPDH:

Glyceraldehyde-3-phosphate dehydrogenase

GLP1RA:

Glucagon-like peptide 1 receptor agonist

GLUT:

Glucose transporter

GPCR:

G protein–coupled receptor

HbA1c:

Hemoglobin A1c

HF:

Heart failure

HFrEF:

Heart failure with reduced ejection fraction

HMGB:

High mobility group box

IKK:

Inhibitor kappa B kinase

IL:

Interleukin

INF:

Interferon

JNK:

c-Jun N-terminal kinase

MACE:

Major adverse cardiovascular event

MAPK:

Mitogen-activated protein kinase

MCP:

Monocyte chemoattractant protein

M-CSF:

Macrophage colony-stimulating factor

MEF:

Myocyte enhancer factor

MRA:

Mineralocorticoid receptor antagonist

NFκB:

Nuclear factor kappa B

NLRP:

NLR family pyrin domain-containing protein

NOX:

Nicotinamide adenine dinucleotide phosphate oxidase

OHP:

Oral hypoglycemic peptide

RAAS:

Renin-angiotensin-aldosterone system

ROS:

Reactive oxygen species

SGLT:

Sodium-glucose cotransporter

TAC:

Transverse aortic constriction

Th:

T helper

TLR:

Toll-like receptor

TNF:

Tumor necrosis factor

3'UTR:

3'-untranslated region

ZDF:

Zucker diabetic fatty

References

  1. Sun H, et al. Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022;183:109119.

    Article  PubMed  Google Scholar 

  2. Standards of medical care in diabetes–2014 Diabetes Care, 2014. 37 Suppl 1: pp. S14-80.

  3. Nichols GA, et al. Congestive heart failure in type 2 diabetes: prevalence, incidence, and risk factors. Diabetes Care. 2001;24(9):1614–9.

    Article  CAS  PubMed  Google Scholar 

  4. Dei Cas A, et al. Impact of diabetes on epidemiology, treatment, and outcomes of patients with heart failure. JACC Heart Fail. 2015;3(2):136–45.

    Article  PubMed  Google Scholar 

  5. Harding JL, et al. Global trends in diabetes complications: a review of current evidence. Diabetologia. 2019;62(1):3–16.

    Article  PubMed  Google Scholar 

  6. Dasari D, et al. Canagliflozin protects diabetic cardiomyopathy by mitigating fibrosis and preserving the myocardial integrity with improved mitochondrial function. Eur J Pharmacol. 2023;949:175720.

    Article  CAS  PubMed  Google Scholar 

  7. Fang P, et al. Glucagon-like peptide-1receptor agonist protects against diabetic cardiomyopathy by modulating microRNA-29b-3p/SLMAP. Drug Des Devel Ther. 2023;17:791–806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yang Z, et al. Metformin ameliorates diabetic cardiomyopathy by activating the PK2/PKR pathway. Front Physiol. 2020;11:425.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Ritchie RH, Abel ED. Basic mechanisms of diabetic heart disease. Circ Res. 2020;126(11):1501–25.

    CAS  Google Scholar 

  10. Phang RJ, et al. Cellular interplay between cardiomyocytes and non-myocytes in diabetic cardiomyopathy. Cardiovasc Res. 2023;119(3):668–90.

    Article  CAS  PubMed  Google Scholar 

  11. Prakoso D, et al. Current landscape of preclinical models of diabetic cardiomyopathy. Trends Pharmacol Sci. 2022;43(11):940–56.

    Article  CAS  PubMed  Google Scholar 

  12. Astorri E, et al. Left ventricular function in insulin-dependent and in non-insulin-dependent diabetic patients: radionuclide assessment. Cardiology. 1997;88(2):152–5.

    Article  CAS  PubMed  Google Scholar 

  13. Yamada T, et al. Sodium-glucose co-transporter-2 inhibitors as add-on therapy to insulin for type 1 diabetes mellitus: systematic review and meta-analysis of randomized controlled trials. Diabetes Obes Metab. 2018;20(7):1755–61.

    Article  CAS  PubMed  Google Scholar 

  14. Mathieu C, et al. Efficacy and safety of liraglutide added to insulin treatment in type 1 diabetes: the ADJUNCT ONE Treat-To-Target Randomized Trial. Diabetes Care. 2016;39(10):1702–10.

    Article  CAS  PubMed  Google Scholar 

  15. Marx N et al. 2023 ESC guidelines for the management of cardiovascular disease in patients with diabetes. Eur Heart J. 2023;44(39):4043–140.

  16. Park MD, et al. Macrophages in health and disease. Cell. 2022;185(23):4259–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122(3):787–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wculek SK, et al. Metabolism of tissue macrophages in homeostasis and pathology. Cell Mol Immunol. 2022;19(3):384–408.

    Article  CAS  PubMed  Google Scholar 

  19. Cassetta L, Cassol E, Poli G. Macrophage polarization in health and disease. ScientificWorldJournal. 2011;11:2391–402.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bashir S, et al. Macrophage polarization: the link between inflammation and related diseases. Inflamm Res. 2016;65(1):1–11.

    Article  CAS  PubMed  Google Scholar 

  21. Peng C, Li Z, Yu X. The role of pancreatic infiltrating innate immune cells in acute pancreatitis. Int J Med Sci. 2021;18(2):534–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sterling KG et al. Mucosal immunity and the Gut-Microbiota-Brain-Axis in Neuroimmune Disease. Int J Mol Sci, 2022. 23(21).

  23. Hu Z, et al. Depletion of macrophages with clodronate liposomes partially attenuates renal fibrosis on AKI-CKD transition. Ren Fail. 2023;45(1):2149412.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Braga TT, Agudelo JS, Camara NO. Macrophages during the fibrotic process: M2 as friend and Foe. Front Immunol. 2015;6:602.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Mills CD, et al. M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 2000;164(12):6166–73.

    Article  CAS  PubMed  Google Scholar 

  26. Locati M, Curtale G, Mantovani A. Diversity, mechanisms, and significance of macrophage plasticity. Annu Rev Pathol. 2020;15:123–47.

    Article  CAS  PubMed  Google Scholar 

  27. Nahrendorf M, Swirski FK. Abandoning M1/M2 for a network model of macrophage function. Circ Res. 2016;119(3):414–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gosselin D, et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell. 2014;159(6):1327–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Tan Y, et al. Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence. Nat Rev Cardiol. 2020;17(9):585–607.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Jia G, Hill MA, Sowers JR. Diabetic Cardiomyopathy: an update of mechanisms contributing to this clinical entity. Circ Res. 2018;122(4):624–38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Seferović PM, Paulus WJ. Clinical diabetic cardiomyopathy: a two-faced disease with restrictive and dilated phenotypes. Eur Heart J. 2015;36(27):1718–27.

    Article  PubMed  Google Scholar 

  32. Marwick TH, et al. Implications of underlying mechanisms for the Recognition and Management of Diabetic Cardiomyopathy. J Am Coll Cardiol. 2018;71(3):339–51.

    Article  PubMed  Google Scholar 

  33. Lin HT et al. (1)H nuclear magnetic resonance (NMR)-Based cerebrospinal fluid and plasma metabolomic analysis in type 2 Diabetic patients and Risk Prediction for Diabetic Microangiopathy. J Clin Med, 2019. 8(6).

  34. DeBerge M, et al. Macrophages in Heart failure with reduced versus preserved ejection fraction. Trends Mol Med. 2019;25(4):328–40.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Jin X, et al. Advanced Glycation End products enhance macrophages polarization into M1 phenotype through activating RAGE/NF-κB pathway. Biomed Res Int. 2015;2015:p732450.

    Article  Google Scholar 

  36. Mishra PK, et al. Diabetic Cardiomyopathy: an immunometabolic perspective. Front Endocrinol (Lausanne). 2017;8:72.

    Article  PubMed  Google Scholar 

  37. Watanabe R et al. Glucose metabolism controls disease-specific signatures of macrophage effector functions. JCI Insight, 2018. 3(20).

  38. Mouton AJ, et al. Obesity, hypertension, and Cardiac Dysfunction: Novel roles of Immunometabolism in Macrophage activation and inflammation. Circ Res. 2020;126(6):789–806.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dinh W, et al. Elevated plasma levels of TNF-alpha and interleukin-6 in patients with diastolic dysfunction and glucose metabolism disorders. Cardiovasc Diabetol. 2009;8:58.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Masters SL, Latz E, O’Neill LA. The inflammasome in atherosclerosis and type 2 diabetes. Sci Transl Med. 2011;3(81):81ps17.

    Article  PubMed  Google Scholar 

  41. Biernacka A, et al. Smad3 Signaling promotes Fibrosis while preserving Cardiac and aortic geometry in obese Diabetic mice. Circ Heart Fail. 2015;8(4):788–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bajpai A, Tilley DG. The role of leukocytes in Diabetic Cardiomyopathy. Front Physiol. 2018;9:1547.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Kokkola R, et al. RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scand J Immunol. 2005;61(1):1–9.

    Article  CAS  PubMed  Google Scholar 

  44. Volz HC, et al. The role of HMGB1/RAGE in inflammatory cardiomyopathy. Semin Thromb Hemost. 2010;36(2):185–94.

    Article  CAS  PubMed  Google Scholar 

  45. Tang SCW, Yiu WH. Innate immunity in diabetic kidney disease. Nat Rev Nephrol. 2020;16(4):206–22.

    Article  CAS  PubMed  Google Scholar 

  46. Frantz S, et al. Toll4 (TLR4) expression in cardiac myocytes in normal and failing myocardium. J Clin Invest. 1999;104(3):271–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhang Y, et al. Prevention of hyperglycemia-induced myocardial apoptosis by gene silencing of toll-like receptor-4. J Transl Med. 2010;8:133.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Dong B, et al. TLR4 regulates cardiac lipid accumulation and diabetic heart disease in the nonobese diabetic mouse model of type 1 diabetes. Am J Physiol Heart Circ Physiol. 2012;303(6):H732–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Tao A, et al. Cardiomyocyte-fibroblast interaction contributes to diabetic cardiomyopathy in mice: role of HMGB1/TLR4/IL-33 axis. Biochim Biophys Acta. 2015;1852(10 Pt A):2075–85.

    Article  CAS  PubMed  Google Scholar 

  50. Wang Y, et al. Saturated palmitic acid induces myocardial inflammatory injuries through direct binding to TLR4 accessory protein MD2. Nat Commun. 2017;8:13997.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Schenk S, Saberi M, Olefsky JM. Insulin sensitivity: modulation by nutrients and inflammation. J Clin Invest. 2008;118(9):2992–3002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest. 2005;115(5):1111–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Aguirre V, et al. The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of ser(307). J Biol Chem. 2000;275(12):9047–54.

    Article  CAS  PubMed  Google Scholar 

  54. Gao Z, et al. Serine phosphorylation of insulin receptor substrate 1 by inhibitor kappa B kinase complex. J Biol Chem. 2002;277(50):48115–21.

    Article  CAS  PubMed  Google Scholar 

  55. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259(5091):87–91.

    Article  CAS  PubMed  Google Scholar 

  56. Weisberg SP, et al. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–808.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Xu H, et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J Clin Invest. 2003;112(12):1821–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219–46.

    Article  CAS  PubMed  Google Scholar 

  59. Patsouris D, et al. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab. 2008;8(4):301–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kanda H, et al. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J Clin Invest. 2006;116(6):1494–505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Uysal KT, et al. Protection from obesity-induced insulin resistance in mice lacking TNF-alpha function. Nature. 1997;389(6651):610–4.

    Article  CAS  PubMed  Google Scholar 

  62. Xu X, et al. Obesity activates a program of lysosomal-dependent lipid metabolism in adipose tissue macrophages independently of classic activation. Cell Metab. 2013;18(6):816–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Jaitin DA, et al. Lipid-Associated Macrophages Control Metabolic Homeostasis in a Trem2-Dependent manner. Cell. 2019;178(3):686–e69814.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Aouadi M, et al. Lipid storage by adipose tissue macrophages regulates systemic glucose tolerance. Am J Physiol Endocrinol Metab. 2014;307(4):E374–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Lafuse WP, Wozniak DJ, Rajaram MVS. Role of Cardiac macrophages on Cardiac inflammation, fibrosis and tissue repair. Cells, 2020. 10(1).

  66. Hulsmans M, Sam F, Nahrendorf M. Monocyte and macrophage contributions to cardiac remodeling. J Mol Cell Cardiol. 2016;93:149–55.

    Article  CAS  PubMed  Google Scholar 

  67. Dick SA, et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol. 2019;20(1):29–39.

    Article  CAS  PubMed  Google Scholar 

  68. Wagner MJ, Khan M, Mohsin S. Healing the broken heart; the Immunomodulatory effects of Stem Cell Therapy. Front Immunol. 2020;11:639.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Horckmans M, et al. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J. 2017;38(3):187–97.

    CAS  PubMed  Google Scholar 

  70. Frati G, et al. An overview of the inflammatory signalling mechanisms in the myocardium underlying the development of diabetic cardiomyopathy. Cardiovasc Res. 2017;113(4):378–88.

    Article  CAS  PubMed  Google Scholar 

  71. Liu G, et al. The effect of mir-471-3p on macrophage polarization in the development of diabetic cardiomyopathy. Life Sci. 2021;268:118989.

    Article  CAS  PubMed  Google Scholar 

  72. Sharifi-Rad M, et al. Lifestyle, oxidative stress, and antioxidants: back and forth in the pathophysiology of Chronic diseases. Front Physiol. 2020;11:694.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Cai L, Kang YJ. Oxidative stress and diabetic cardiomyopathy: a brief review. Cardiovasc Toxicol. 2001;1(3):181–93.

    Article  CAS  PubMed  Google Scholar 

  74. Wilson AJ, et al. Reactive oxygen species signalling in the diabetic heart: emerging prospect for therapeutic targeting. Heart. 2018;104(4):293–9.

    Article  CAS  PubMed  Google Scholar 

  75. Nishikawa T, et al. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature. 2000;404(6779):787–90.

    Article  CAS  PubMed  Google Scholar 

  76. Cai L, et al. Inhibition of superoxide generation and associated nitrosative damage is involved in metallothionein prevention of diabetic cardiomyopathy. Diabetes. 2005;54(6):1829–37.

    Article  CAS  PubMed  Google Scholar 

  77. Zhu HZ, et al. Xiaoyaosan exerts therapeutic effects on the Colon of Chronic Restraint stress model rats via the regulation of Immunoinflammatory Activation Induced by the TLR4/NLRP3 Inflammasome Signaling Pathway. Evid Based Complement Alternat Med. 2021;2021:p6673538.

    Google Scholar 

  78. Pal PB, et al. Aldose Reductase mediates NLRP3 inflammasome-initiated Innate Immune Response in Hyperglycemia-Induced Thp1 monocytes and male mice. Endocrinology. 2017;158(10):3661–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Peng ML, et al. Signaling pathways related to oxidative stress in Diabetic Cardiomyopathy. Front Endocrinol (Lausanne). 2022;13:907757.

    Article  PubMed  Google Scholar 

  80. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res. 2010;107(9):1058–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes. 2005;54(6):1615–25.

    Article  CAS  PubMed  Google Scholar 

  82. Yan SD, et al. Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J Biol Chem. 1994;269(13):9889–97.

    Article  CAS  PubMed  Google Scholar 

  83. Rendra E, et al. Reactive oxygen species (ROS) in macrophage activation and function in diabetes. Immunobiology. 2019;224(2):242–53.

    Article  CAS  PubMed  Google Scholar 

  84. Gerstein HC, et al. Long-term effects of intensive glucose lowering on cardiovascular outcomes. N Engl J Med. 2011;364(9):818–28.

    Article  CAS  PubMed  Google Scholar 

  85. Chalmers J, Cooper ME. UKPDS and the legacy effect. N Engl J Med. 2008;359(15):1618–20.

    Article  CAS  PubMed  Google Scholar 

  86. Wang T, et al. Influenza-trained mucosal-resident alveolar macrophages confer long-term antitumor immunity in the lungs. Nat Immunol. 2023;24(3):423–38.

    Article  CAS  PubMed  Google Scholar 

  87. Netea MG, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol. 2020;20(6):375–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Edgar L, et al. Hyperglycemia induces trained immunity in macrophages and their precursors and promotes atherosclerosis. Circulation. 2021;144(12):961–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Wood IS, Trayhurn P. Glucose transporters (GLUT and SGLT): expanded families of sugar transport proteins. Br J Nutr. 2003;89(1):3–9.

    Article  CAS  PubMed  Google Scholar 

  90. Xie L, et al. Emerging roles of Sodium glucose cotransporter 2 (SGLT-2) inhibitors in Diabetic Cardiovascular diseases: focusing on immunity, inflammation and metabolism. Front Pharmacol. 2022;13:836849.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Lee SH, Park SY, Choi CS. Insulin resistance: from mechanisms to therapeutic strategies. Diabetes Metab J. 2022;46(1):15–37.

    Article  PubMed  Google Scholar 

  92. Shaffner J, et al. Therapeutic targeting of SGLT2: a new era in the treatment of Diabetes and Diabetic kidney disease. Front Endocrinol (Lausanne). 2021;12:749010.

    Article  PubMed  Google Scholar 

  93. Kato ET, et al. Effect of Dapagliflozin on Heart failure and mortality in type 2 diabetes Mellitus. Circulation. 2019;139(22):2528–36.

    Article  CAS  PubMed  Google Scholar 

  94. Furtado RHM, et al. Dapagliflozin and Cardiovascular outcomes in patients with type 2 diabetes Mellitus and previous myocardial infarction. Circulation. 2019;139(22):2516–27.

    Article  CAS  PubMed  Google Scholar 

  95. Lee TM, Chang NC, Lin SZ. Dapagliflozin, a selective SGLT2 inhibitor, attenuated cardiac fibrosis by regulating the macrophage polarization via STAT3 signaling in infarcted rat hearts. Free Radic Biol Med. 2017;104:298–310.

    Article  CAS  PubMed  Google Scholar 

  96. Iannantuoni F et al. The SGLT2 inhibitor Empagliflozin ameliorates the Inflammatory Profile in type 2 Diabetic patients and promotes an antioxidant response in leukocytes. J Clin Med, 2019. 8(11).

  97. Kim SR, et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat Commun. 2020;11(1):2127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wu Q, et al. Dapagliflozin protects against chronic heart failure in mice by inhibiting macrophage-mediated inflammation, independent of SGLT2. Cell Rep Med. 2023;4(12):101334.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Xu L, et al. SGLT2 inhibition by Empagliflozin Promotes Fat Utilization and Browning and attenuates inflammation and insulin resistance by polarizing M2 macrophages in Diet-induced obese mice. EBioMedicine. 2017;20:137–49.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Xu L, Ota T. Emerging roles of SGLT2 inhibitors in obesity and insulin resistance: focus on fat browning and macrophage polarization. Adipocyte. 2018;7(2):121–8.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Terami N, et al. Long-term treatment with the sodium glucose cotransporter 2 inhibitor, dapagliflozin, ameliorates glucose homeostasis and diabetic nephropathy in db/db mice. PLoS ONE. 2014;9(6):e100777.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Oelze M, et al. The sodium-glucose co-transporter 2 inhibitor empagliflozin improves diabetes-induced vascular dysfunction in the Streptozotocin diabetes rat model by interfering with oxidative stress and glucotoxicity. PLoS ONE. 2014;9(11):e112394.

    Article  PubMed  PubMed Central  Google Scholar 

  103. McMurray JJV, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381(21):1995–2008.

    Article  CAS  PubMed  Google Scholar 

  104. Wiviott SD, et al. Dapagliflozin and Cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380(4):347–57.

    Article  CAS  PubMed  Google Scholar 

  105. Zinman B, et al. Empagliflozin, Cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117–28.

    Article  CAS  PubMed  Google Scholar 

  106. Neal B, et al. Canagliflozin and Cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377(7):644–57.

    Article  CAS  PubMed  Google Scholar 

  107. Ye Y, et al. SGLT-2 inhibition with Dapagliflozin reduces the activation of the Nlrp3/ASC inflammasome and attenuates the Development of Diabetic Cardiomyopathy in mice with type 2 diabetes. Further augmentation of the effects with Saxagliptin, a DPP4 inhibitor. Cardiovasc Drugs Ther. 2017;31(2):119–32.

    Article  PubMed  Google Scholar 

  108. Kern M, et al. The SGLT2 inhibitor empagliflozin improves insulin sensitivity in db/db mice both as monotherapy and in combination with linagliptin. Metabolism. 2016;65(2):114–23.

    Article  CAS  PubMed  Google Scholar 

  109. Habibi J, et al. Sodium glucose transporter 2 (SGLT2) inhibition with empagliflozin improves cardiac diastolic function in a female rodent model of diabetes. Cardiovasc Diabetol. 2017;16(1):9.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Orskov C, et al. Tissue and plasma concentrations of amidated and glycine-extended glucagon-like peptide I in humans. Diabetes. 1994;43(4):535–9.

    Article  CAS  PubMed  Google Scholar 

  111. MacDonald PE, et al. The multiple actions of GLP-1 on the process of glucose-stimulated insulin secretion. Diabetes. 2002;51(Suppl 3):S434–42.

    Article  CAS  PubMed  Google Scholar 

  112. Lazzaroni E, et al. Anti-diabetic drugs and weight loss in patients with type 2 diabetes. Pharmacol Res. 2021;171:105782.

    Article  CAS  PubMed  Google Scholar 

  113. Kristensen SL, et al. Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol. 2019;7(10):776–85.

    Article  CAS  PubMed  Google Scholar 

  114. Bethel MA, et al. Cardiovascular outcomes with glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes: a meta-analysis. Lancet Diabetes Endocrinol. 2018;6(2):105–13.

    Article  PubMed  Google Scholar 

  115. Pop-Busui R, et al. Heart failure: an underappreciated complication of diabetes. A Consensus Report of the American Diabetes Association. Diabetes Care. 2022;45(7):1670–90.

    Article  PubMed  PubMed Central  Google Scholar 

  116. Almutairi M, et al. The GLP-1 receptor agonist Liraglutide increases myocardial glucose oxidation Rates via Indirect mechanisms and mitigates Experimental Diabetic Cardiomyopathy. Can J Cardiol. 2021;37(1):140–50.

    Article  PubMed  Google Scholar 

  117. Trang NN et al. Empagliflozin and Liraglutide differentially modulate Cardiac Metabolism in Diabetic Cardiomyopathy in rats. Int J Mol Sci, 2021. 22(3).

  118. Qian P, et al. A novel oral glucagon-like peptide 1 receptor agonist protects against diabetic cardiomyopathy via alleviating cardiac lipotoxicity induced mitochondria dysfunction. Biochem Pharmacol. 2020;182:114209.

    Article  CAS  PubMed  Google Scholar 

  119. Ma X, et al. GLP-1 receptor agonists (GLP-1RAs): cardiovascular actions and therapeutic potential. Int J Biol Sci. 2021;17(8):2050–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Wan S, Sun H. Glucagon-like peptide-1 modulates RAW264.7 macrophage polarization by interfering with the JNK/STAT3 signaling pathway. Exp Ther Med. 2019;17(5):3573–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Wang N, et al. Exendin-4 induces bone marrow stromal cells Migration through Bone Marrow-Derived macrophages polarization via PKA-STAT3 signaling pathway. Cell Physiol Biochem. 2017;44(5):1696–714.

    Article  CAS  PubMed  Google Scholar 

  122. Yang L, et al. Effect of GLP-1/GLP-1R on the polarization of macrophages in the occurrence and development of atherosclerosis. Mediators Inflamm. 2021;2021:p5568159.

    Article  Google Scholar 

  123. Weisberg SP, et al. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest. 2006;116(1):115–24.

    Article  CAS  PubMed  Google Scholar 

  124. Winer S, et al. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med. 2009;15(8):921–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Lee YS, et al. Glucagon-like peptide-1 inhibits adipose tissue macrophage infiltration and inflammation in an obese mouse model of diabetes. Diabetologia. 2012;55(9):2456–68.

    Article  CAS  PubMed  Google Scholar 

  126. Tate M, et al. Exendin-4 attenuates adverse cardiac remodelling in streptozocin-induced diabetes via specific actions on infiltrating macrophages. Basic Res Cardiol. 2016;111(1):1.

    Article  CAS  PubMed  Google Scholar 

  127. Gerstein HC, et al. The effect of dulaglutide on stroke: an exploratory analysis of the REWIND trial. Lancet Diabetes Endocrinol. 2020;8(2):106–14.

    Article  CAS  PubMed  Google Scholar 

  128. Tuttolomondo A, et al. Efficacy of dulaglutide on vascular health indexes in subjects with type 2 diabetes: a randomized trial. Cardiovasc Diabetol. 2021;20(1):1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Marso SP, et al. Liraglutide and Cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375(4):311–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Husain M, et al. Oral Semaglutide and Cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2019;381(9):841–51.

    Article  CAS  PubMed  Google Scholar 

  131. Strain WD, et al. Effects of Semaglutide on Stroke subtypes in type 2 diabetes: Post Hoc Analysis of the Randomized SUSTAIN 6 and PIONEER 6. Stroke. 2022;53(9):2749–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Gerstein HC, et al. Cardiovascular and renal outcomes with Efpeglenatide in Type 2 diabetes. N Engl J Med. 2021;385(10):896–907.

    Article  CAS  PubMed  Google Scholar 

  133. Hernandez AF, et al. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (harmony outcomes): a double-blind, randomised placebo-controlled trial. Lancet. 2018;392(10157):1519–29.

    Article  CAS  PubMed  Google Scholar 

  134. van der Aa MP, et al. Long-term treatment with metformin in obese, insulin-resistant adolescents: results of a randomized double-blinded placebo-controlled trial. Nutr Diabetes. 2016;6(8):e228.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Giannarelli R, et al. Reducing insulin resistance with metformin: the evidence today. Diabetes Metab. 2003;29(4 Pt 2):s628–35.

    Google Scholar 

  136. Xiao H, et al. Metformin attenuates cardiac fibrosis by inhibiting the TGFbeta1-Smad3 signalling pathway. Cardiovasc Res. 2010;87(3):504–13.

    Article  CAS  PubMed  Google Scholar 

  137. Hippisley-Cox J, Coupland C. Diabetes treatments and risk of heart failure, cardiovascular disease, and all cause mortality: cohort study in primary care. BMJ. 2016;354:i3477.

    Article  PubMed  PubMed Central  Google Scholar 

  138. Crowley MJ, et al. Clinical outcomes of Metformin Use in populations with chronic kidney disease, congestive heart failure, or Chronic Liver Disease: a systematic review. Ann Intern Med. 2017;166(3):191–200.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Nassif RM et al. Metformin inhibits ROS production by human M2 macrophages via the activation of AMPK. Biomedicines, 2022. 10(2).

  140. Sánchez ML et al. Polymers and Bioactive compounds with a macrophage modulation effect for the Rational Design of Hydrogels for skin regeneration. Pharmaceutics, 2023. 15(6).

  141. Das B et al. Promise of the NLRP3 inflammasome inhibitors in in vivo Disease models. Molecules, 2021. 26(16).

  142. Horiuchi T, et al. Metformin directly binds the alarmin HMGB1 and inhibits its proinflammatory activity. J Biol Chem. 2017;292(20):8436–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Feng X, et al. Metformin, macrophage dysfunction and atherosclerosis. Front Immunol. 2021;12:682853.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Guo C, Liu J, Li H. Metformin ameliorates olanzapine-induced insulin resistance via suppressing macrophage infiltration and inflammatory responses in rats. Biomed Pharmacother. 2021;133:110912.

    Article  CAS  PubMed  Google Scholar 

  145. Cortés M, et al. Inflammatory macrophages reprogram to immunosuppression by reducing mitochondrial translation. Nat Commun. 2023;14(1):7471.

    Article  PubMed  PubMed Central  Google Scholar 

  146. Panagiotopoulou O, et al. Metformin use in obese mothers is associated with improved cardiovascular profile in the offspring. Am J Obstet Gynecol. 2020;223(2):246.e1–10.

    Article  Google Scholar 

  147. Timmons JG, et al. Metformin and carotid intima-media thickness in never-smokers with type 1 diabetes: the REMOVAL trial. Diabetes Obes Metab. 2021;23(6):1371–8.

    Article  CAS  PubMed  Google Scholar 

  148. Eurich DT, et al. Comparative safety and effectiveness of metformin in patients with diabetes mellitus and heart failure: systematic review of observational studies involving 34,000 patients. Circ Heart Fail. 2013;6(3):395–402.

    Article  CAS  PubMed  Google Scholar 

  149. Bhansali S, et al. Metformin upregulates mitophagy in patients with T2DM: a randomized placebo-controlled study. J Cell Mol Med. 2020;24(5):2832–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Yang F, et al. Metformin inhibits the NLRP3 Inflammasome via AMPK/mTOR-dependent effects in Diabetic Cardiomyopathy. Int J Biol Sci. 2019;15(5):1010–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Tsoyi K, et al. Metformin inhibits HMGB1 release in LPS-treated RAW 264.7 cells and increases survival rate of endotoxaemic mice. Br J Pharmacol. 2011;162(7):1498–508.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Fiordaliso F, et al. Cardiovascular oxidative stress is reduced by an ACE inhibitor in a rat model of streptozotocin-induced diabetes. Life Sci. 2006;79(2):121–9.

    Article  CAS  PubMed  Google Scholar 

  153. Matsusaka H, et al. Angiotensin II type 1 receptor blocker attenuates exacerbated left ventricular remodeling and failure in diabetes-associated myocardial infarction. J Cardiovasc Pharmacol. 2006;48(3):95–102.

    Article  CAS  PubMed  Google Scholar 

  154. Wichter T, et al. Pregnancy in arrhythmogenic cardiomyopathy. Herzschrittmacherther Elektrophysiol. 2021;32(2):186–98.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Sowers JR, Epstein M, Frohlich ED. Diabetes, hypertension, and cardiovascular disease: an update. Hypertension. 2001;37(4):1053–9.

    Article  CAS  PubMed  Google Scholar 

  156. Yang J, et al. Valsartan preconditioning protects against myocardial ischemia-reperfusion injury through TLR4/NF-kappaB signaling pathway. Mol Cell Biochem. 2009;330(1–2):39–46.

    Article  CAS  PubMed  Google Scholar 

  157. Yaras N, et al. Restoration of diabetes-induced abnormal local Ca2 + release in cardiomyocytes by angiotensin II receptor blockade. Am J Physiol Heart Circ Physiol. 2007;292(2):H912–20.

    Article  CAS  PubMed  Google Scholar 

  158. Solomon SD, et al. Effect of the direct renin inhibitor aliskiren, the angiotensin receptor blocker losartan, or both on left ventricular mass in patients with hypertension and left ventricular hypertrophy. Circulation. 2009;119(4):530–7.

    Article  CAS  PubMed  Google Scholar 

  159. Shah AM, et al. Left ventricular systolic and diastolic function, remodelling, and clinical outcomes among patients with diabetes following myocardial infarction and the influence of direct renin inhibition with aliskiren. Eur J Heart Fail. 2012;14(2):185–92.

    Article  CAS  PubMed  Google Scholar 

  160. Nagase M, et al. Salt-induced nephropathy in obese spontaneously hypertensive rats via paradoxical activation of the mineralocorticoid receptor: role of oxidative stress. Hypertension. 2007;50(5):877–83.

    Article  CAS  PubMed  Google Scholar 

  161. Lambert JM, Lopez EF, Lindsey ML. Macrophage roles following myocardial infarction. Int J Cardiol. 2008;130(2):147–58.

    Article  PubMed  PubMed Central  Google Scholar 

  162. Mansour AA, et al. The interplay of galectins-1, -3, and– 9 in the immune-inflammatory response underlying cardiovascular and metabolic disease. Cardiovasc Diabetol. 2022;21(1):253.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Camici M, et al. Obesity-related glomerulopathy and podocyte injury: a mini review. Front Biosci (Elite Ed). 2012;4(3):1058–70.

    PubMed  Google Scholar 

  164. Hansson L, et al. Effect of angiotensin-converting-enzyme inhibition compared with conventional therapy on cardiovascular morbidity and mortality in hypertension: the Captopril Prevention Project (CAPPP) randomised trial. Lancet. 1999;353(9153):611–6.

    Article  CAS  PubMed  Google Scholar 

  165. Yusuf S, et al. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. N Engl J Med. 2000;342(3):145–53.

    Article  CAS  PubMed  Google Scholar 

  166. Lambers Heerspink HJ, et al. Moderation of dietary sodium potentiates the renal and cardiovascular protective effects of angiotensin receptor blockers. Kidney Int. 2012;82(3):330–7.

    Article  PubMed  Google Scholar 

  167. Lefkowitz RJ. A brief history of G-protein coupled receptors (Nobel lecture). Angew Chem Int Ed Engl. 2013;52(25):6366–78.

    Article  CAS  PubMed  Google Scholar 

  168. Milligan G, Svoboda P, Brown CM. Why are there so many adrenoceptor subtypes? Biochem Pharmacol. 1994;48(6):1059–71.

    Article  CAS  PubMed  Google Scholar 

  169. Taylor MR. Pharmacogenetics of the human beta-adrenergic receptors. Pharmacogenomics J. 2007;7(1):29–37.

    Article  CAS  PubMed  Google Scholar 

  170. Xing G, et al. Recent progress in the development of β2 adrenergic receptor agonists: a patent review (2015–2020). Expert Opin Ther Pat. 2021;31(3):239–46.

    Article  CAS  PubMed  Google Scholar 

  171. Ortega E, Gálvez I, Martín-Cordero L. Adrenergic regulation of macrophage-mediated Innate/Inflammatory responses in obesity and Exercise in this Condition: role of β2 adrenergic receptors. Endocr Metab Immune Disord Drug Targets. 2019;19(8):1089–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Noh H, et al. Beta 2-adrenergic receptor agonists are novel regulators of macrophage activation in diabetic renal and cardiovascular complications. Kidney Int. 2017;92(1):101–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Zhang H, et al. Renal-protective effect of thalidomide in streptozotocin-induced diabetic rats through anti-inflammatory pathway. Drug Des Devel Ther. 2018;12:89–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Grisanti LA, et al. β2-Adrenergic receptor-dependent chemokine receptor 2 expression regulates leukocyte recruitment to the heart following acute injury. Proc Natl Acad Sci U S A. 2016;113(52):15126–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Kaumann A, et al. Activation of beta2-adrenergic receptors hastens relaxation and mediates phosphorylation of phospholamban, troponin I, and C-protein in ventricular myocardium from patients with terminal heart failure. Circulation. 1999;99(1):65–72.

    Article  CAS  PubMed  Google Scholar 

  176. Gong J, et al. Caveolin-3 protects diabetic hearts from acute myocardial infarction/reperfusion injury through β2AR, cAMP/PKA, and BDNF/TrkB signaling pathways. Aging. 2020;12(14):14300–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. van Beek SMM, et al. Prolonged β(2)-adrenergic agonist treatment improves glucose homeostasis in diet-induced obese UCP1(-/-) mice. Am J Physiol Endocrinol Metab. 2021;320(3):E619–28.

    Article  PubMed  Google Scholar 

  178. Mahtal N, et al. MicroRNAs in kidney injury and disease. Nat Rev Nephrol. 2022;18(10):643–62.

    Article  CAS  PubMed  Google Scholar 

  179. Guo X, et al. miR-335 negatively regulates osteosarcoma stem cell-like properties by targeting POU5F1. Cancer Cell Int. 2017;17:29.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Duroux-Richard I, Apparailly F, Khoury M. Mitochondrial MicroRNAs contribute to Macrophage Immune functions including differentiation, polarization, and activation. Front Physiol. 2021;12:738140.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Zhong Y, Yi C. MicroRNA-720 suppresses M2 macrophage polarization by targeting GATA3. Biosci Rep, 2016. 36(4).

  182. Ying H, et al. MiR-127 modulates macrophage polarization and promotes lung inflammation and injury by activating the JNK pathway. J Immunol. 2015;194(3):1239–51.

    Article  CAS  PubMed  Google Scholar 

  183. Nazari-Jahantigh M, et al. MicroRNA-155 promotes atherosclerosis by repressing Bcl6 in macrophages. J Clin Invest. 2012;122(11):4190–202.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Li D, et al. MiR-146a modulates macrophage polarization in systemic juvenile idiopathic arthritis by targeting INHBA. Mol Immunol. 2016;77:205–12.

    Article  CAS  PubMed  Google Scholar 

  185. Muñoz JP, et al. The transcription factor MEF2C mediates cardiomyocyte hypertrophy induced by IGF-1 signaling. Biochem Biophys Res Commun. 2009;388(1):155–60.

    Article  PubMed  Google Scholar 

  186. Ruiz MA, Chakrabarti S. MicroRNAs: the underlying mediators of pathogenetic processes in vascular complications of diabetes. Can J Diabetes. 2013;37(5):339–44.

    Article  PubMed  Google Scholar 

  187. Moore A, et al. Rapid onset of cardiomyopathy in STZ-induced female diabetic mice involves the downregulation of pro-survival Pim-1. Cardiovasc Diabetol. 2014;13:68.

    Article  PubMed  PubMed Central  Google Scholar 

  188. Kuwabara Y, et al. MicroRNA-451 exacerbates lipotoxicity in cardiac myocytes and high-fat diet-induced cardiac hypertrophy in mice through suppression of the LKB1/AMPK pathway. Circ Res. 2015;116(2):279–88.

    Article  CAS  PubMed  Google Scholar 

  189. Liu S, et al. Micro-RNA 21Targets dual specific phosphatase 8 to promote collagen synthesis in high glucose-treated primary cardiac fibroblasts. Can J Cardiol. 2014;30(12):1689–99.

    Article  PubMed  Google Scholar 

  190. Yildirim SS, et al. Relationship between downregulation of miRNAs and increase of oxidative stress in the development of diabetic cardiac dysfunction: junctin as a target protein of miR-1. Cell Biochem Biophys. 2013;67(3):1397–408.

    Article  CAS  PubMed  Google Scholar 

  191. Wang P, et al. miR-657 promotes macrophage polarization toward M1 by targeting FAM46C in gestational diabetes Mellitus. Mediators Inflamm. 2019;2019:p4851214.

    Article  Google Scholar 

  192. Zhuang G, et al. A novel regulator of macrophage activation: miR-223 in obesity-associated adipose tissue inflammation. Circulation. 2012;125(23):2892–903.

    Article  CAS  PubMed  Google Scholar 

  193. Fitzsimons S, et al. microRNA-155 is decreased during atherosclerosis regression and is increased in urinary extracellular vesicles during atherosclerosis progression. Front Immunol. 2020;11:576516.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Jia C, et al. Gold nanoparticle-based miR155 antagonist macrophage delivery restores the cardiac function in ovariectomized diabetic mouse model. Int J Nanomed. 2017;12:4963–79.

    Article  CAS  Google Scholar 

  195. Phu TA, et al. IL-4 polarized human macrophage exosomes control cardiometabolic inflammation and diabetes in obesity. Mol Ther. 2022;30(6):2274–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Bouchareychas L, et al. Macrophage exosomes resolve atherosclerosis by regulating hematopoiesis and inflammation via MicroRNA Cargo. Cell Rep. 2020;32(2):107881.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Li JM, et al. Lipotoxicity-polarised macrophage-derived exosomes regulate mitochondrial fitness through Miro1-mediated mitophagy inhibition and contribute to type 2 diabetes development in mice. Diabetologia. 2023;66(12):2368–86.

    Article  CAS  PubMed  Google Scholar 

  198. Sun J, et al. miR-330-5p/Tim-3 axis regulates macrophage M2 polarization and insulin resistance in diabetes mice. Mol Immunol. 2018;95:107–13.

    Article  CAS  PubMed  Google Scholar 

  199. Cao J, et al. Extracellular vesicle miR-32 derived from macrophage promotes arterial calcification in mice with type 2 diabetes via inhibiting VSMC autophagy. J Transl Med. 2022;20(1):307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Chang YJ, et al. Extracellular MicroRNA-92a mediates endothelial cell-macrophage communication. Arterioscler Thromb Vasc Biol. 2019;39(12):2492–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Zhang M, et al. MiR-130b promotes obesity associated adipose tissue inflammation and insulin resistance in diabetes mice through alleviating M2 macrophage polarization via repression of PPAR-γ. Immunol Lett. 2016;180:1–8.

    Article  CAS  PubMed  Google Scholar 

  202. Suresh Babu S, et al. MicroRNA-126 overexpression rescues diabetes-induced impairment in efferocytosis of apoptotic cardiomyocytes. Sci Rep. 2016;6:36207.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Acuña-Castroviejo D, et al. Extrapineal melatonin: sources, regulation, and potential functions. Cell Mol Life Sci. 2014;71(16):2997–3025.

    Article  PubMed  Google Scholar 

  204. Nabavi SM, et al. Anti-inflammatory effects of melatonin: a mechanistic review. Crit Rev Food Sci Nutr. 2019;59(sup1):S4–16.

    Article  CAS  PubMed  Google Scholar 

  205. Stebelová K, Herichová I, Zeman M. Diabetes induces changes in melatonin concentrations in peripheral tissues of rat. Neuro Endocrinol Lett. 2007;28(2):159–65.

    PubMed  Google Scholar 

  206. Fiorina P, et al. Impaired nocturnal melatonin excretion and changes of immunological status in ischaemic stroke patients. Lancet. 1996;347(9002):692–3.

    Article  CAS  PubMed  Google Scholar 

  207. Maity J, et al. Melatonin ameliorates myocardial infarction in obese diabetic individuals: the possible involvement of macrophage apoptotic factors. J Pineal Res. 2023;74(2):e12847.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Nicole Stancel, PhD, ELS(D), and Shiladitya Sengupta, PhD, of the Department of Scientific Publications at The Texas Heart Institute contributed to the editing of this review article.

Funding

This research was funded by the Special Foundation Projects of Joint Applied Basic Research of Yunnan Provincial Department of Science and Technology with Kunming Medical University, grant number 202301AY070001-119; the Natural Science Foundation, grant number 82270372; the Program Innovative Research Team in Science and Technology in Kunming Medical University, grant number 202405AS350014; and the Yunnan Health Training Project of High-Level Talents, grant number H-2019052.

Author information

Authors and Affiliations

Authors

Contributions

C.Y.Z., A.S., and Y.K.S. conceived and designed the review; C.Y.Z. and Y.K.S. drafted and wrote the manuscript; A.S., S.M.S., Q.L., P.Y.L., and H.Y.C. reviewed and revised the manuscript; C.Y.Z., C.Z.L., Z.H., and Y.M.M. composed the figures and tables. All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Ao Shi or Hongyan Cai.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, C., Shi, Y., Liu, C. et al. Therapeutic strategies targeting mechanisms of macrophages in diabetic heart disease. Cardiovasc Diabetol 23, 169 (2024). https://doi.org/10.1186/s12933-024-02273-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12933-024-02273-4

Keywords