Abstract
Chronic kidney disease (CKD) is a serious and progressive worldwide health problem affecting 15% of the global population. CKD is associated to greater mortality rates due to secondary complications such as cardiovascular disease. Usual cardiovascular complications found in CKD patients include left ventricular hypertrophy, heart failure, and cardiac arrhythmias. The most common type of cardiac arrhythmia in CKD patients is atrial fibrillation (AF). Proper management of AF is important due to its high risk of cardiovascular complications and stroke. The incidence of AF remains higher in CKD patients than in the healthy population, highlighting the need for improving our understanding of the mechanisms underlying CKD-induced AF. In this review, we discussed well-known systemic factors linking CKD to AF pathogenesis. We highlighted the involvement of several inflammatory mediators in the CKD-induced atrial arrhythmogenesis. We also addressed special considerations for experimental models of CKD and the management of AF in CKD patients. Finally, we emphasize the need for a deeper understanding of the molecular underpinning, and for high-quality clinical investigations into the CKD-AF connection.
Keywords: Atrial fibrillation, calcium signaling, chronic kidney disease, inflammation, oxidative stress
Graphical Abstract

Potential CKD-induced inflammatory effects on left atria responsible of atrial fibrillation (AF). We highlighted how kidney dysfunction induces a cytokine storm that in turn stimulates bone marrow cytokine production and travel to the heart where it stimulates left atrial dilatation, electrical, and structural remodeling in cardiomyocytes, causes activation of fibroblasts, and recruitment of macrophages, creating a substrate for the development of atrial fibrillation (AF). CM: cardiomyocyte; CF: cardiac fibroblast; FGF-23: fibroblast growth factor 23; IL-1β: interleukin 1 beta; IL-6: interleukin 6; IL-17: interleukin 17; IL-18: interleukin 18; RyR2: ryanodine receptor type 2; STAT3: signal transducer and activator of transcription 3; TGFβ: transforming growth factor beta; TNFα: tumor necrosis factor alpha.
1. INTRODUCTION
Chronic kidney disease (CKD), a pathology characterized by the progressive loss of kidney function, is affecting 10–15% of the global population [1], and is responsible for significant morbidity and mortality [2, 3]. The ‘Kidney Disease: Improving Global Outcomes (KDIGO) Guidelines’ defined CKD as abnormal kidney structure or function for at least 3 months with implications for health [4]. Cardiovascular disease is responsible of almost half of all the deaths in CKD patients [5]. Common cardiac events in CKD patients are left ventricular hypertrophy (LVH), heart failure (HF) [6], ischemic heart disease, vascular calcification [7], and arrhythmias [8, 9]. Among cardiac complications, atrial fibrillation (AF) is the most common arrhythmia in CKD patients with an estimate rate of 37.6 episodes of AF per person-year [10, 11]. AF is a supraventricular tachyarrhythmia characterized by rapid and uncoordinated atrial electrical activation and ineffective atrial contraction. Clinically, AF is classified based on the duration of episodes and whether it self-terminates within 7 days: paroxysmal AF (terminates spontaneously within 7 days), persistent AF (lasts more than 7 days), and permanent AF (long-lasting and refractory to rhythm control) [12]. The initiation and maintenance of AF is due to two major mechanisms, ectopic electrical activity (triggered activity) and reentry because of structural and/or electrical remodeling, respectively [13]. The aberrant Ca2+ handling that can lead to early or delayed after depolarizations (EADs and DADs, respectively) or enhanced automaticity are the primary causes of triggered activity. Electrical remodeling plays a key role in reentry and to the maintenance of AF by shortening of the action potential duration (APD) due to altered activities of repolarizing ion channels. On the other hand, structural remodeling, characterized by atrial fibrosis and enlargement, impairs conduction and promotes the heterogeneity in electrical conduction, facilitating the maintenance of reentry [14]. In this review, we examine the clinical evidence of the heightened association between CKD and AF development, discuss the mechanisms linking CKD and AF pathogenesis, and highlight special considerations for AF management in CKD patients. We also provide our perspectives on future research opportunities in this area.
2. Heightened Association Between AF and CKD
It has been reported that AF occurs in approximately one in five non-dialysis CKD patients [15] and one in three dialysis-dependent CKD patients [16]. AF incidence rises as kidney function declines, with more than a two-fold increase in CKD patients whose estimated glomerular filtration rate (eGFR) is below 30 ml/min/1.73 m2 compared to those with an eGFR above 60 ml/min/1.73m2 [17]. A recent meta-analysis has shown that kidney dysfunction increases the risk of AF by almost 65% compared to healthy controls [18]. Among adults younger than 55 years, the rate of AF was more than 10 times higher in CKD patients compared to the general population (females: 1.9% vs. 0.1%; males: 2.5% vs. 0.2%) [16]. In the United States, 18–22% of CKD patients develop AF. Patients who suffer from both CKD and AF pathologies have a worse prognosis than those with only one of these conditions. CKD patients who develop AF have an elevated risk of stroke, heart failure, myocardial infraction, kidney dysfunction progression, and all-cause mortality [19–21]. The interrelatedness between AF and CKD is, in part, caused by shared risk factors, such as hypertension, heart failure, vascular disease, diabetes mellitus (DM), and aging [22]. Nevertheless, recent studies have proposed that CKD itself serves as an independent risk factor for new onset AF [23, 24]. Studies have shown that both paroxysmal and non-paroxysmal AF are more prevalent in CKD patients. Electrolyte imbalances during dialysis have been associated with paroxysmal AF episodes [25], while chronic CKD-induced oxidative stress and inflammation have been linked to persistent AF in CKD patients [26]. Furthermore, CKD-induced systemic alterations often lead to cardiac fibrosis, which disrupts electrical conduction in the heart and increases the risk of developing persistent AF [27]. However, it remains unclear whether improved kidney function in CKD patients can reverse atrial remodeling, particularly in those with persistent AF.
The CKD-associated AF risk differs by sex. Studies have shown that males with CKD have a higher risk of AF development than females with CKD, partly due to the protective effect of estrogens and deleterious effects of testosterone in CKD patients [28, 29]. Recent studies indicate that testosterone deficiency is common in CKD patients, and male hypogonadism may both impact and be influences by CKD [30]. On the other hand, in the menopausal woman, the ovarian senescence and the loss of estrogen are directly associated with a decline in renal function and indirectly with an accumulation of cardiometabolic risk factors [31].
3. Molecular alterations linking CKD and AF Pathogenesis
The increased cardiovascular risk associated with CKD is largely driven by systemic changes, such as the accumulation of uremic toxins, electrolyte imbalances, and neurohormonal dysregulation. These factors contribute to tissue and systemic oxidative stress, activation of the renin-angiotensin-aldosterone system (RAAS), and inflammation [32], which in turn are key mechanisms underlying the development of a pro-arrhythmic substrate. Below, we discuss common pathological changes that are associated with CKD development including hyperglycemia, RAAS, mineral bone disorders, and oxidative stress (Figure 1, Table 1). We also highlight the inflammatory cytokines associated with AF pathogenesis in the context of CKD.
Figure 1. Intracellular pathways activated in cardiomyocytes in response to chronic kidney disease (CKD) that might lead to atrial fibrillation (AF).

Important pathways activated in cardiomyocytes from CKD patients in response to high glucose levels, angiotensin II, hyperphosphatemia, FGF-23 and PTH, and inhibited pathways due to low levels of vitamin D and Klotho, that might lead to structural or electrical remodeling in atrial cardiomyocytes. 1,25-OH-VitD: 1,25-dihidroxy-vitamin D; AC: adenyl cyclase; AMPK: AMP-activated protein kinase; AngII: angiotensin II; ANP: atrial natriuretic peptide; AT-II: angiotensin receptor II; ATP: adenosin-triphosphate; BNP: brain natriuretic peptide; Ca2+: calcium ion; CaMKII: calmodulin kinase type II; cAMP: cyclic adenosin-monophosphate; ETRA/B: endothelin receptor A/B; FGF-23: fibroblast growth factor 23; FGFR4: FGF receptor 4; GLUT1/4: glucose transporter 1/4; IP3: inositol-triphosphate; IP3R: IP3-receptor; JAK2: janus kinase 2; LTCC: L-type calcium channel; MHC: major histocompatibility complex; mTOR: mammalian target of rapamycin; NOX2: NADPH oxidase 2; NFAT: nuclear factor of activated T cell; Nrf2: nuclear factor erythroid 2-related factor 2; Pi: phosphate; PIP3: phosphor-inositol triphosphate; Pit1/2: phosphate transporter ½; PKA: protein kinase A; PLCγ: phospholipase C gamma; PLN: phospholamban; PP1: protein phosphatase 1; PTH: parathyroid hormone; PTHR: PTH receptor; Rcan: regulator of calcineurin gene; ROS: reactive oxygen species; RYR2: ryanodine receptor type 2; SERCA2a: sarcoplasmic reticulum Ca2+ ATPase 2a; Sirt3: sirtuin 3; SOD2: superoxide dismutase 2; STAT3: signal transducer and activator of transcription 3; TGFβ: transforming growth factor beta; TRPC: transient receptor potential cation channel subfamily C; VDR: vitamin D receptor.
Table 1.
Key Mediators Altered in Chronic Kidney Disease and Their Reported Arrhythmogenic Effects
| Mediators | Status in CKD | Reported arrhythmogenesis effects |
|---|---|---|
| Glucose | Increased | • Enlargement of left atria [37] • Cardiomyocyte apoptosis [38] • Mitochondrial dysfunction [39] • Hyperactivity of RyR2 channel [40] |
| Angiotensin | Increased | • Increased atrial fibrosis [53] • Decreased SERCA2a activity [52] • Enhanced CaMKII oxidation [51] |
| Phosphate | Increased | • Vascular calcification [61] • Hyperphosphorylation of RyR2 and enhanced activation of STAT3 [65] |
| Vitamin D | Decreased | • Increased cardiac fibrosis [74] |
| PTH | Increased | • Increased Ca2+ current [83] • Cardiomyocyte hypertrophy [83] |
| FGF-23 | Increased | • Cardiomyocyte hypertrophy [88] • Hyperactivity of RyR2 channel, increased SCaEs, and contractile dysfunction [91] |
| ROS | Increased | • CaMKII hyperactivity [102] • Increased late INa,L [104] |
| IL-6 | Increased | • Increased cardiac fibrosis [116] • Enhanced activation of STAT3 [115] |
| TNFα | Increased | • Atrial fibrosis [137] • Decreased SERCA2a activity [138] • Contractile dysfunction [138] |
| IL-1β/IL-18 | Increased | • Hyperphosphorylation of RyR2 [142] • Enlargement of left atria [152] |
| IL-17 | Increased | • Increased atrial fibrosis [167] |
| NLRP3 | Increased | • Enlargement of left atria, atrial fibrosis, ectopic activity [156] |
| TGF-β | Increased | • Increased atrial fibrosis [198] |
3.1. Hyperglycemia
Diabetes mellitus (DM) and CKD are two of the strongest risk factors for developing cardiovascular complications [33], including cardiac arrhythmias. DM is a metabolic disorder characterized by persistent elevations of serum glucose levels. DM is known to independently increase AF risk and further exacerbate it in CKD patients [34]. Interestingly, DM is also a known risk factor for kidney disease. It is estimated that almost 40% of DM patients develop CKD [35], commonly known as diabetic nephropathy (DN). forrelationship between DM and CKD, as kidney disease can exacerbate DM by impairing glucose filtration and reabsorption, which leads to insulin resistance and further elevates blood glucose [36]. Glucose resistance in DM has been associated with glycogen granule accumulation in the myocardium, which leads to stiffness of cardiac tissue and left atrial (LA) enlargement [37], an established pro-AF substrate. The diabetic milieu can directly affect cardiac muscle contractility by inducing cardiomyocyte apoptosis and malformation of contractile structures [38]. Recent studies also show that high glucose levels induce cardiomyocyte mitochondrial dysfunction mediated by endothelin-1 (ET-1) receptor activation and the AMPK-mTOR pathway [39]. High glucose levels can directly activate G-proteins that increase calcium influx into cardiomyocytes via the L-type calcium channels, and subsequently induce the calcium release via the ryanodine receptor type-2 (RyR2) [40], increasing the propensity for pro-AF triggered activity. Moreover, DM is also associated with inflammation and oxidative stress [41], whose effects on AF are discussed later in this review.
3.2. Renin-angiotensin-aldosterone system (RAAS)
CKD promotes hypertension through mechanisms like fluid retention and increased plasma volume, along with the local activation of the RAAS in response to renal injury. This activation increases the production of angiotensin II (Ang II), a known vasopressor. RAAS activation plays an important role in the intertwining progression of both CKD and hypertension [42]. Notably, hypertension is a common risk factor of AF, with over 70% of cases reporting this comorbidity [43, 44]. Though the molecular mechanisms connecting hypertension and AF pathogenesis are multifactorial, it is widely accepted that hypertension leads to structural remodeling of the atria via either hemodynamic changes or RAAS activation [43]. Consistently, atrial cardiomyocytes of AF patients exhibit elevated levels angiotensin converting enzyme (ACE) and angiotensin II receptors (AT-II) compared to patients in sinus rhythm [45], while RAAS antagonists have been shown to reduce atrial fibrosis [46, 47] and electrical remodeling [48], leading to a lower recurrence of AF in patients with paroxysmal AF [49]. Activation of angiotensin receptor 2 (AT-II) activates the JAK-STAT pathway leading to the expression of important pro-hypertrophic genes [50].
RAAS activation during CKD can contribute to both triggered activities and electrical/structural remodeling. On one hand, Ang II has been linked to AF development independently of kidney function. The Ang II infusion model has been shown to induce AF as a result of increased atrial fibrosis, electrical remodeling, and inflammation [51]. Ang II increases levels of NADPH oxidase 2 (NOX2) in cardiomyocytes, driving the production of reactive oxygen species (ROS) [52]. Excess ROS can evoke abnormal calcium release and decrease SERCA activity by impeding the dephosphorylation of phospholamban (PLN) through inhibition of protein phosphatase 1 (PP1) or by oxidizing CaMKII, thereby promoting triggered activity and arrhythmogenesis [51]. Furthermore, recent studies have identified platelet activation as a mediator of Ang II effects. Platelets conjugate with fibroblasts and activates them by secreting TGF-β1, which promotes atrial fibrosis and AF [53]. On the other hand, during the CKD-associated hypertension, pressure overload-induced atrial enlargement not only impairs electrical conduction but also facilitates the formation of reentrant circuits within the atria. Furthermore, pressure overload can induce macrophage infiltration, which in turn affects the functions of cardiomyocytes and cardiac fibroblasts through paracrine signaling mechanisms [54, 55].
3.3. Mineral Bone Disorders
Mineral bone disorders (MBD) consist of mineral disturbances closely associated with CKD [56]. MBD includes altered serum calcium concentrations, increased serum phosphate levels (hyperphosphatemia), augmented parathyroid hormone (PTH) levels, reduced active vitamin D, increased fibroblast growth factor (FGF)-23, and decreased levels of the antiaging protein Klotho [57]. Alterations in mineral homeostasis are known to increase the risk of cardiovascular disease by augmenting atherosclerosis and vascular calcification [58]. The latter has been found to increase mortality in CKD patients [59]. MBDs have been demonstrated to be strongly associated with AF development in CKD patients, especially for FGF-23 [60]
3.3.1. Phosphate
It is well known that electrolyte imbalances, including high phosphate levels, can disrupt normal cardiac rhythm, leading to arrhythmias. Hyperphosphatemia is a significant cardiovascular risk factor and shows the strongest correlation to vascular calcification in CKD patients [61]. Increased coronary artery calcium score has been associated to increased risk of AF development [62]. Consistent with this, several large population-based studies have found an association between higher phosphate levels and an increased incidence of AF [63, 64]. Hsu et al. recently demonstrated that hyperphosphatemia in a 5/6 nephrectomy animal model induced atrial remodeling and increased the AF incidence by activating STAT3/NF-κB signaling and oxidative stress [65]. Exposure of myocytes to high phosphate levels in vitro induced hyperphosphorylation of RyR2, which may predispose to the increased incidence of arrhythmias [66]. Furthermore, reduced phosphorus excretion due to renal dysfunction induces secondary hyperparathyroidism and increases serum FGF-23 levels, contributing to additional cardiovascular complications.
3.3.2. Vitamin D
CKD patients often exhibit low serum vitamin D levels [67] due to reduced activity of 1α-hydroxylase CYP27B1, an enzyme responsible for converting inactive vitamin D to its active form, along with compromised skin xerosis, dietary restrictions, and loss of vitamin D binding proteins due to proteinuria [68]. Data from National Health and Nutrition Examination Survey (NHANES) and the National Death Index (NDI) (2007‒2018) database show that low vitamin D levels in CKD patients are associated to higher cardiovascular disease-related mortality [69]. Observational studies have suggested that vitamin D deficiency is linked to an increased risk of AF development even in general population [70]. A recent 5-year follow-up study in a large cohort of healthy population showed that the risk of new onset AF is reduced in those supplemented with vitamin D [71]. Additionally, CKD patients with AF have significantly lower calcidiol (25-hydroxy-vitamin D) levels compared with those without AF [72]. This protective effect of vitamin D might be due to the reduction in cardiac fibrosis mediated by the activation of the vitamin D receptors (VDRs). It has been shown that VRD activation suppressed preproendothelin gene expression in cardiac fibroblast [73]. In this sense, it has been shown that mice lacking VRDs showed increased cardiac fibrosis [74]. Activation of VRDs in CKD rats has been also shown to reduce LVH and cardiac fibrosis [75, 76] reducing the risk of developing AF. Furthermore, it has been shown that activation of VDR activates the nuclear factor erythroid 2-related factor 2 (Nrf2) promoting the expression of sirtuin-3 (Sirt3) which induces SOD2 expression in the mitochondria to protect against ROS [77].
3.3.3. Parathyroid hormone (PTH)
Increased serum phosphate levels due to kidney dysfunction stimulate the parathyroid glands, leading to increased PTH synthesis, a condition known as secondary hyperparathyroidism. The National Kidney Foundation’s Kidney Disease Outcomes Quality Initiative (KDOQI) guideline highlight that PTH levels rise as kidney function declines [78]. Although the Atherosclerosis Risk in Communities (ARIC) study failed to show any association between PTH levels and HF, peripheral artery disease and cardiovascular mortality [79], recent findings from the PTH Target Trial show that reducing PTH levels can significantly lower mortality and cardiovascular complications in CKD patients [80]. Additionally, PTH levels correlate with left atrial enlargement [81], and have been found to be higher in patients with AF compared to healthy population [82], being further elevated in persistent AF compared to those with paroxysmal AF. Mechanistically, activation of PTH1R, a receptor for PTH, in cardiomyocytes triggers calcium influx and PLC-PKC pathway activation, promoting cell proliferation and cardiac hypertrophy in rats [83]. PTH also increases cAMP levels in cardiomyocytes, activating PKA which phosphorylates RyR2 increasing calcium leak that might lead to cardiac arrhythmias [84]. Furthermore, lowering the PTH concentrations improves cell shortening by increasing calcium load in isolated cardiomyocytes from rats [85].
3.3.4. FGF-23
Phosphate dysregulation in CKD increases the expression of the phosphaturic hormone FGF-23 expression, leading to elevated systemic FGF-23 levels. Serum FGF-23 levels have been found to be 1000-fold higher in late-stage CKD compared to the healthy population [86]. Under normal conditions, FGF-23 promotes kidney phosphate excretion. However, pathological elevations in FGF-23 levels are correlated with cardiovascular complications. FGF-23 has been shown to cause LVH and cardiovascular mortality, particularly in CKD patients [87]. It induces cardiac hypertrophy in a kotho-independent FGFR4-mediated mechanism by activating the PLCγ-calcineurin-NFAT signaling pathway [88, 89]. Activation of PLC increases the levels of inositol-3-phosphate (IP3), activating the IP3 receptors (IP3R) that releases calcium from sarcoplasmic reticulum activating calcineurin that promotes the expression of pro-hypertrophic genes by activation of the nuclear factor of activated T cells (NFAT). FGF-23 has been widely associated with prevalent and incident AF in patients with CKD [60, 90]. This association with AF might be due to increased incidence of LVH but also electrophysiological changes in cardiomyocytes. Thus, FGF-23 has also been shown to cause contractile dysfunction and intracellular calcium mishandling in cardiomyocytes, even in the absence of kidney disease [87, 91]. However, Pastor-Arroyo et al. found that FGF-23 alone, without kidney disease, does not induce any cardiovascular alterations [92]. Together, this evidence suggests that other CKD-induced factors may work synergistically with FGF-23 to promote AF pathogenesis.
3.3.5. Klotho
Klotho, known as an anti-aging protein, is mainly secreted by the kidneys and binds to FGF-23 receptor 1 (FGFR1), increasing the receptor’s affinity for FGF-23 to promote phosphate excretion. Klotho levels decrease as kidney disease progresses [86]. The incidence of AF is higher in CKD patients with lower serum Klotho levels [93, 94] and klotho levels are significantly smaller in dialysis-dependent CKD patients with AF compared to those in sinus rhythm [93]. However, more studies are needed to elucidate if only a Klotho reduction without increasing FGF-23 levels is associated to increased incidence of AF or whether that is a casual association. It has been shown that klotho prevents fibrosis by inhibiting PLC pathway in atrial fibroblast and suppressing TRP currents [95] that might confer protection against AF even in the CKD milieu.
3.4. Oxidative Stress
Oxidative stress results from an imbalance between the production of ROS and the antioxidant capacity, which leads to oxidative damage to protein, lipids, and DNA. Oxidative stress is a major mediator of the cardiovascular diseases, including CKD and AF [96]. CKD is associated with both overproduction of ROS and a reduction of antioxidant defense activity (catalase, glutathione peroxidase, and glutathione). Pro-oxidants activity is usually a consequence of CKD risk factors, such as age, diabetes, chronic inflammation, uremic toxins, and dialysis treatment [97]. Besides Ang-II induced NOX2 activation discussed earlier, CKD is linked to increased levels of oxidized low-density lipoprotein (LDL) [98], xanthine oxidase (XOD) [99], and indoxyl sulfate (IS)-mediated oxidative stress [100], as well as reduced levels of antioxidants SOD and GSH [101]. Increased ROS has been shown to directly affect CaMKII activity [102], a key protein kinase regulating intracellular calcium handling in cardiomyocytes. ROS-dependent CaMKII activation occurs via oxidation at methionines 281 and 282 by NADPH oxidase [103]. Oxidized CaMKII can promote AF pathogenesis by enhancing the late sodium current (INa,L) or increased diastolic calcium leak through increased RyR2 phosphorylation [51, 104]. For further details on oxidative stress in atrial arrhythmogenesis, readers are referred to another expert review in this issue [105].
3.5. Inflammation in CKD and AF
Chronic systemic inflammation is a central driver of AF in CKD [106]. It is well documented that that systemic inflammation correlates with the AF development in patients. C-reactive protein (CRP) levels, an indicator of systemic inflammation, increase in a dose-dependent manner with AF burden [107]. Circulating cytokines, key modulators of immune and inflammatory response [108], can exert a wide range of effects on various organ systems such as the heart [109] and kidney. Within the atria, the elevated levels of inflammatory cytokines can promote atrial remodeling and adrenergic hyperactivation, both of which promote AF [110, 111]. In this section, we discuss key inflammatory cytokines that have been mechanistically linked to AF pathogenesis in the context of CKD (Figure 2, Table 1).
Figure 2. Intracellular pathways activated in cardiomyocytes by CKD-induced interleukins that may cause atrial fibrillation (AF).

Key pathways activated in response to CKD-induced increases in inflammatory cytokines that lead to electrical and structural remodeling in atria. ATA2a2: sarcoplasmic reticulum Ca2+-ATPase 2a gene; ASC: apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD); Ca2+: calcium ion; CACNA1c: Ca2+ voltage-gated channel subunit alpha 1c gene; CaMKII: Ca2+/calmodulin-dependent protein kinase type II; GSDM: gasdermin D; gp130: glycoprotein 130; IL-1β: interleukin 1 beta; IL1R: IL-1 receptor; IL-6: interleukin 6; IL6Rα: IL-6 receptor α; IL-17: interleukin 17; IL17R: IL-17 receptor; IL-18: interleukin 18; JAK2: janus kinase 2; NF-kB: nuclear factor kappa-B; NLRP3: nucleotide-binding oligomerization domain (NOD)-, leucin-rich repeat protein (LRR)- and pyridine domain-containing protein 3; PLN: phospholamban; ROS: reactive oxygen species; RYR2: ryanodine receptor type 2; SERCA2a: sarcoplasmic reticulum Ca2+ ATPase 2a; STAT3: signal transducer and activator of transcription 3; TNFα: tumor necrosis factor alpha; TNFR: TNFβ receptor.
3.5.1. Interleukin (IL)-6
IL-6 belongs to a family of cytokines including IL-11 and cardiotrophin-1 that signal through a common transmembrane receptor glycoprotein 130 (gp130) [112]. IL-6 signaling can be pro- or anti-inflammatory, depending on the manner of signaling [113]. Classical IL-6 signaling is anti-inflammatory and involves binding of IL-6 to gp130 and membrane-bound IL-6 receptor alpha (IL-6Ra), which is only expressed on leukocytes and hepatocytes [113]. In contrast, trans-signaling is pro-inflammatory and involves binding of IL-6 to soluble IL-6Ra, which is cleaved from leukocytes via ADAM17 [114], and then to gp130 [113]. Thus, pro-inflammatory IL-6 trans-signaling occurs in all cells including cardiomyocytes, except for those that express IL-6Ra [113]. Regardless of the mode of IL-6 signaling, IL-6 activation results in STAT3 dimerization and subsequent transcriptional upregulation of its target genes [112]. Unlike IL-6Ra, IL-6 is secreted by all cell types [113]. CKD can increase IL-6 levels through a variety of mechanisms. CKD can activate the RAAS, mediated through angiotensin II, which can activate STAT3 in cardiomyocytes and cardiac fibroblasts via the AT1 receptor, increasing IL-6 synthesis [115]. Angiotensin II-induced IL-6 synthesis has been shown to be a key mediator of angiotensin II-induced cardiac hypertrophy and fibrosis [116]. Lastly, sympathetic nervous system activation in CKD [117] can also induce IL-6 synthesis [118].
Clinical studies have demonstrated a strong association between plasma IL-6 levels and renal function in CKD. A longitudinal study of 899 CKD individuals from the Chronic Renal Insufficiency Cohort (CRIC) found that baseline plasma IL-6 levels correlated with GFR decline over a median follow-up of 6.3 years [119]. Importantly, the association between IL-6 and GFR decline remained significant after adjustment for key risk factors including baseline GFR, age, sex, race, cholesterol, hypertension, diabetes, RAAS inhibitor therapy, body mass index, alcohol use, and smoking, whereas the association between IL-1β and GFR decline became nonsignificant [119] – indicating that IL-6 is a specific and direct mediator of CKD pathophysiology. With regard to AF in CKD, a cohort study of 3,762 CKD patients showed that baseline plasma IL-6 was independently associated with prevalent and new-onset AF over a mean follow-up of 3.7 years [120]. Indeed, plasma IL-6 levels are predictive of all-cause and cardiovascular mortality in CKD patients [121]. Moreover, a phase 2 randomized controlled trial comparing anti-IL-6 monoclonal antibody to placebo in hemodialysis patients demonstrated that IL-6 inhibition decreased inflammation-driven anemia of chronic disease [122], in conjunction with decreased markers of systemic inflammation such as CRP and fibrinogen [123]. These results indicate that targeting the IL-6 signaling axis in CKD has the potential to abrogate deleterious complications of CKD. However, it remains unclear whether anti-IL-6 monoclonal antibody can also reduce AF incidence in CKD patients.
Mechanistically, CKD-mediated IL-6 upregulation can lead to AF through a variety of mechanisms, one of which is via IL-6-mediated FGF-23 upregulation [124]. As discussed earlier, in CKD, FGF-23 is upregulated in a compensatory manner to combat hyperphosphatemia by inhibiting proximal tubular phosphate resorption and decreasing parathyroid gland parathyroid hormone secretion [125, 126]. However, upregulated FGF-23 may also be detrimental for AF development in CKD patients [60]. Within the atria, FGF-23 has been shown to promote atrial remodeling and fibrosis [127], and IL-6 was shown to increase FGF-23 in a STAT3-dependent manner [124]. Indeed, global knockout of Il-6 in adenine-high-phosphorus diet-induced CKD mice prevented pathologic increases in circulating FGF-23 and rescued CKD-associated hypocalcemia. Taken together, CKD-induced IL-6 upregulation can lead to AF through IL-6-mediated FGF-23 upregulation and resulting FGF-23-mediated atrial remodeling, which is discussed in more detail within the mineral bone disorders section of this review.
Fibrosis is another key mechanism by which IL-6 leads to AF in CKD. Administration of soluble gp130, which selectively inhibits pro-inflammatory IL-6 trans-signaling [113], abrogates STAT3-induced renal fibrosis and kidney function deterioration in a unilateral ureteral occlusion mouse model [128]. In the heart, IL-6 also promotes atrial fibrosis and AF in a STAT3-dependent manner and inhibition of STAT3 with S3I-201 decreases AF inducibility in rats [129]. Lastly, IL-6 has been shown to directly promote arrhythmogenic changes in calcium handling as IL-6 perfusion of isolated rat hearts increased time to peak calcium transient amplitude and susceptibility to calcium transient alternans [50].
3.5.2. Tumor necrosis factor (TNF)-α
TNF-α, a central mediator of systemic inflammation, is the only other cytokine (in addition to IL-6) that predicts risk of new-onset AF in CKD patients [130]. Serum TNF-α levels are positively correlated with histologic kidney damage, indicating that it plays a direct role in CKD pathophysiology [131]. Indeed, a study of rats that underwent renal reduction surgery to induce CKD demonstrated that systemic administration of a soluble TNF-α neutralizing receptor (PEG-sTNFR1) for six weeks ameliorated CKD-related hypertension, renal inflammation and fibrosis, and albuminuria [132]. TNF-α neutralization in mice with diabetic nephropathy has also been shown to protect against renal dysfunction and ameliorate CKD progression [133], indicating that TNF-α mediates CKD pathology of various etiologies. Interestingly, macrophage-specific TNF-α conditional knockout in a mouse model of diabetic nephropathy reduced albuminuria and histologic evidence of renal damage, demonstrating that TNF-α produced by macrophages is sufficient to recapitulate its systemic pathologic effects in CKD [133].
With regard to AF, serum TNF-α is elevated in AF compared to sinus rhythm patients [134], and a study of human atrial tissue showed that intra-atrial TNF-α and IL-6 levels were greater in valvular disease patients with AF compared to those without AF [135]. Indeed, treating human atrial cardiac fibroblasts in vitro with TNF-α increases release of IL-6 [136], indicating that these two cytokines function in tandem. Mechanistically, TNF-α can promote atrial structural and electrical remodeling. Recombinant TNF-α administered via tail vein injection to mice was sufficient to increase atrial fibrosis and reduce connexin-40 [137] – substrate alterations that promote reentry. Consistently, cardiac-specific TNF-α overexpression resulted in atrial dilatation and spontaneous AF, with prolonged calcium reuptake [138]. Notably, transgenic TNF-α overexpression is one of only four documented mouse models of spontaneous AF [139], with AF starting at five months-of-age at 77% incidence [138, 139]. Altogether, there is strong preclinical and clinical evidence that elevated TNF-α in CKD directly promotes AF through atrial structural and electrical remodeling.
3.5.3. IL-1β/IL-18
Unlike IL-6 and TNF-α, IL-1β and IL-18 show a weaker association in clinical studies of CKD-related AF. A post hoc analysis of the REDHART (Recently Decompensated Heart Failure Anakinra Response Trial) found no benefit of Anakinra, an IL-1β neutralizing monoclonal antibody, in preventing decline of renal function in patients with cardiorenal syndrome, although patients in the Anakinra treatment group exhibited decreased markers of systemic inflammation such as CRP and myeloperoxidase [140]. Similarly, the CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcomes Study) trial, a double-blind randomized controlled trial comparing IL-1β inhibition via canakinumab versus placebo in patients after myocardial infarction with high-sensitivity CRP of at least 2 mg per liter, showed no improvement of renal function despite a reduction in overall cardiovascular disease risk in the treatment group [141]. Nonetheless serum IL-1β are elevated in CKD patients [119] prior to multivariate adjustment, and elevated serum IL-1β has been shown to lead to AF by augmenting calcium mishandling [142]. Acute IL-1β application to HL-1 atrial cardiomyocytes in vitro increases arrhythmogenic calcium release events, likely secondary to RyR2-Ser2814 phosphorylation, which increases RyR2 channel open probability and arrhythmogenic diastolic calcium leak [142, 143]. IL-1β also induces IL-6 synthesis [144], and thus increased IL-1β can lead to AF through mechanisms related to IL-6 discussed above. In line with this, anti-inflammatory agent colchicine was shown to abrogate AF inducibility in rats through inhibition of IL-1β-induced IL-6 synthesis and subsequent IL-6-mediated atrial fibrosis [145].
IL-18 belongs to the IL-1β family and is produced primarily by macrophages and monocytes. However, renal IL-18 expression is upregulated in renal pathology, particularly diabetic nephropathy [146]. IL-18 levels show a strong positive correlation with albuminuria and renal disease progression in diabetic nephropathy [146]. In the heart, IL-18 can lead to AF through a variety of mechanisms, including induction of IL-1β synthesis [147, 148], oxidative stress [149], and recruitment of immune cells through upregulation of cellular adhesion molecules [150, 151]. Indeed, circulating IL-18 levels are elevated in AF patients compared to sinus rhythm controls, with greater levels seen with increasing AF burden and correlating with left atrial diameter [152, 153]. Furthermore, a study performed on 87 AF patients undergoing cryoablation showed that those with persistent or permanent AF had higher IL-18 levels in the left atrial blood compared to paroxysmal AF patients, and that levels in the left atrial blood were higher than those in the venous blood, indicating that IL-18 plays a key role in local atrial inflammation [154]. Indeed, intra-atrial Il-18 gene expression was upregulated in a mouse model of postoperative atrial fibrillation [155]. In a recent study, we show that serum level of IL-1β was increased in hemodialysis patients with AF compared with those in sinus rhythm [156]. Because most of studies to date show that a role of IL-1β and IL-18 in CKD-related AF may be weaker than that for IL-6 and TNF-α, we speculate that IL-1β and IL-18 likely mediate AF progression in CKD, albeit in tandem with other cytokines and mechanisms discussed throughout this review.
3.5.4. IL-17
IL-17 has been implicated in CKD pathogenesis [157]. IL-17 production is increased in Th17 cells in end-stage renal disease patients compared to healthy controls [158], and a separate study of hemodialysis patients found that the percentage of circulating Th17 cells correlated with serum phosphate levels, indicating that Th17 cells are key mediators of renal damage in CKD [159]. Indeed, IL-17 has been shown to accurately predict severity of kidney damage in lupus nephritis with a C-statistic of 0.91 [160]. Importantly, the IL-17 signaling axis is promoted by IL-6 via skewing of naïve T-cell differentiation towards a Th17 phenotype [161]. In the heart, IL-17 signaling is increased in AF patients compared to sinus rhythm controls [162]. While the same study did not find a correlation between serum IL-17 and AF burden, there was a strong positive correlation between IL-17 and neutrophil to lymphocyte ratio, indicating that IL-17 may perpetuate macrophage-mediated inflammation in AF [163]. Furthermore, a rat model of chronic AF induced by 28 days of acetylcholine and CaCl2 tail vein injection found upregulation of IL-17 signaling pathways in AF compared to sham rats [164]. As discussed above, IL-17 can promote monocyte recruitment [165], which is implicated in AF pathogenesis [163], and forms a positive feedback, self-amplifying loop with IL-6 [166], which is involved in AF pathophysiology as discussed above. IL-17 may also promote atrial fibrotic remodeling as anti-IL-17 monoclonal antibody treatment of rats was shown to ameliorate atrial fibrosis in a rat sterile pericarditis model of postoperative atrial fibrillation [167].
3.5.5. IL-10
Unlike IL-17, IL-10 is considered an anti-inflammatory cytokine [168]. Genetic polymorphisms in the IL-10 gene that increase serum IL-10 levels protect against diabetic nephropathy [169]. A study of rats that underwent 5/6 nephrectomy to induce CKD found that IL-10 overexpression via administration of a recombinant adeno-associated virus serotype 1 (AAV1) IL-10 vector attenuated renal inflammation, interstitial fibrosis, proteinuria, and GFR decline [170]. Consistently, administration of IL-10 in mice fed a high-fat diet for 12 weeks reduced fibrotic and inflammatory atrial remodeling as well as AF inducibility [171]. Similar findings were observed in rat model of AF induced by pressure overload, where depletion of spleen-derived IL-10 increased AF susceptibility secondary to inflammation-mediated atrial fibrosis [172]. While the evidence is strong that IL-10 mechanistically protects against CKD and AF progression, studies linking CKD-mediated dysregulation of IL-10 signaling in AF pathogenesis are lacking.
3.5.6. Inflammasomes
Inflammasomes are a key component of the innate immune system that has been shown to play a critical role in the pathogenies and development of AF [173]. Inflammasomes are multiprotein complexes composed of receptor proteins, adaptor proteins, and caspases that regulate cytokine maturation, inflammation, and cell death. Formation of functional inflammasome can be initiated by pattern recognition receptors (PRRs), which can sense pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). PRRs that can assemble inflammasomes include the nucleotide-binding oligomerization domain (NOD), leucine-rich repeat (LRR)-containing protein (NLR) family members NLRP1, NLRP3, and NLRC4, as well as the proteins absent in melanoma 2 (AIM2) and toll-like receptor (TLR). Assembly of the inflammasome components involves the interaction of inflammasome receptors with the apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and effector protein pro-caspase-1 (canonical pathway). This activates the effector caspase through proteolytic cleavage. The activated caspase cleaves full-length pro-IL-18 and pro-IL-1β, producing their active forms [174].
Increased activity of atrial NLRP3 inflammasome has been linked to the development of AF in patients [174, 175]. Various studies have investigated the role of the NLRP3 inflammasome in various models of kidney disease, such as 5/6 nephrectomy, acute kidney injury (AKI), and hypertension-related renal damage, among others [176–178]. It has been reported that nephrectomy-induced CKD rats developed left atrial enlargement, interstitial fibrosis, and pacing-induced AF through the activation of TGFβ-1/Smad2/3 and NLRP3 inflammasome signaling [179]. Consistently, we recently show that genetic ablation of NLRP3 attenuates electrical remodeling, prevents atrial fibrosis, and reduces AF susceptibility in the 5/6 nephrectomy-induced CKD model [156]. The effect of NLRP3 inflammasome activation on CKD-associated AF is primarily mediated by IL-1β, and the increased circulating IL-1β in CKD mice promotes a positive feedforward relationship with the activity of NLRP3 inflammasome in atria. Administration of an IL-1β neutralizing antibody in CKD mice not only prevents atrial enlargement, atrial fibrosis, and AF susceptibility, but also reduces the activity of inflammasome in atria of CKD mice [156].
AIM2 is a cytoplasmic sensor of double-stranded DNA (dsDNA) from pathogens, including bacteria, viruses, fungi, and parasites [180]. AIM2 also responds to dsDNA released from damaged cellular organelles, resulting in the secretion of downstream cytokines, thereby promoting sterile inflammatory diseases such as skin disease, neuronal disease, CKD, and CVD [181]. AIM2 is expressed in kidney and can be activated in the diabetic or nondiabetic CKD patients [182]. In a renal injury model induced by the unilateral urethral obstruction, genetic ablation of AIM2 ameliorates renal injury, fibrosis, and inflammation [182]. This is accompanied by reduced levels of the inflammasome effector cytokines IL-β and IL-18 in the kidney. It has been suggested that AIM2 inflammasome recognizes the necrotic DNA and subsequently drives the IL-1β and IL-18 release from macrophages in the kidney. The role of AIM2 in the cardiovascular diseases has been documented, including coronary atherosclerosis [183], myocardial infarction [184], and myocarditis [185], and heart failure [186]. While direct evidence of AIM2 in CKD associated AF is currently lacking, we recently showed that AIM2 plays a role in the high-protein-diet induced AF by promoting mitochondrial dysfunction [187].
3.5.7. Transforming growth factor beta (TGF-β)
TGF-β is a key mediator of fibrosis, and cardiac-specific overexpression of TGF-β1 in mice was sufficient to cause atrial fibrosis and inducible AF [188, 189]. Strikingly, whole heart cardiomyocyte-specific TGF-β1 transgenic overexpression driven by the myosin heavy chain promoter selectively affects the atria without substantial alterations in ventricular fibrosis [190], suggesting that atria may be particularly sensitive to the pro-fibrotic effects of TGF-β1. TGF-β1 can promote fibrosis in the kidneys [191]. Elevated serum TGF-β levels in type II diabetic patients are associated with greater risk of nephropathy [192]. However, the effects of TGF-β1 inhibition are complicated by the fact that the latent form of TGF-β1 can protect renal function. Transgenic mice with overexpression of latent TGF-β1 are protected against renal inflammation and fibrosis after unilateral ureteral obstruction [193]. Most clinical trials have failed to show that systemic TGF-β neutralization can rescue the GFR decline in CKD [194–196], perhaps owing to the fact that most circulating TGF-β1 is the latent (i.e., protective) form [197]. Targeting pathologic pathways downstream of TGF-β1 signaling may bypass this effect. For instance, neutralization of TGF-β1 downstream mediator SMAD3 via small molecule inhibitor petchiether A in a mouse model of obstructive uropathy protected against renal fibrosis [198]. Overall, TGF-β1 plays a role in pathologic fibrotic and inflammatory remodeling in the kidneys and atria, although a direct link between the two organs remains elusive. One possibility is that the elevated levels of circulating TGF-β1 in CKD can promote atrial fibrosis, creating a substrate for AF development. Another possibility is that fibrocytes, which are circulating fibroblast progenitor cells, can mediate crosstalk between the kidney and atria, particularly in relation to TGF-β1 [199]. Nonetheless, further investigations are needed to clarify how TGF-β1-mediated fibrosis in the kidney can lead to atrial fibrosis and AF.
4. Experimental models of CKD
To date, a substantial body of work has studied the CKD in animal models. Table 2 highlights several commonly used murine models of CKD, which are established through methods that induce renal mass reduction, glomerular injury, nephropathy, nephritis, or polycystic kidney disease, among others [200, 201]. Most studies examining the cardiac consequences of kidney dysfunction in these models have primarily been focusing on the ventricular function and structure. Notably, the 5/6 nephrectomy (also known as subtotal nephrectomy model) is the most used model for studying AF pathogenesis in kidney diseases. In this model, atrial fibrosis and inflammation, key contributors to AF pathogenesis, are consistently reproduced [156, 179]. While each model has its advantages and disadvantages, the choice of CKD model should be guided by the following criteria: 1) the targeted stage of CKD, 2) surgical reproducibility, 3) the rate and progression of CKD, and 4) the specific context of CKD development (e.g., polycystic kidney disease).
Table 2.
Comparison of Various Murine Models of Chronic Kidney Disease.
| Model | Cardiac outcomes | Advantages | Disadvantages |
|---|---|---|---|
| 5/6 nephrectomy [156, 178] | • Cardiac hypertrophy • Cardiac fibrosis • Increased inflammation • Atrial enlargement |
• Well-stablished model • Mimics the late stages of CKD • Reliable CKD induction |
• Surgery induced |
| Unilateral nephrectomy [233, 234] | • Cardiac hypertrophy • Cardiac fibrosis |
• High survival rate • Mimics early stages of CKD • Reliable CKD induction |
• Surgery induced • Low reproducibility |
| Renal ischemia/reperfusion injury [235, 236] | • Cardiac fibrosis • Cardiac hypertrophy |
• Mimics early stages of CKD • Reliable CKD induction |
• Surgery induced • High mortality rate • Slower progression to CKD |
| Adenine diet [237, 238] | • Cardiac fibrosis • Cardiac hypertrophy • Increased inflammation |
• Diet induced • Reliable CKD induction |
• Limited relevance to humans |
| Oxalate nephropathy [239, 240] | • Cardiac fibrosis | • Diet induced • Recapitulate common CKD complications • Reliable CKD induction |
• Possible cardiac toxicity • Limited relevance to humans |
| Alport mice [241] | • Cardiac fibrosis • Cardiac hypertrophy • Increased inflammation |
• Genetic model | • Slower progression to CKD (19 – 21 weeks) |
| Polycystic kidney disease [242] | • Cardiac hypertrophy | • Genetic model | • Translational relevance limited to polycystic kidney disease patients |
5. AF Management in CKD
The main treatment for CKD patients involves renal replacement therapy, including dialysis and renal transplantation. AF incidence has been reported to be nearly 30% in dialysis-dependent CKD patients [202] and around 7% in patients undergoing kidney transplantation within the first 3 years post-transplant [203]. Additionally, AF incidence in kidney transplant recipients is associated with post-transplant mortality [204], which underscores the critical need for improved AF management in CKD patients. However, medication in CKD patients presents unique challenges due to impaired kidney function affecting drug metabolism and excretion. While rhythm control is a standard therapeutic strategy, selecting appropriate antiarrhythmic drugs requires caution. Water-soluble agents such as sotalol, which is renally excreted, should be avoided in CKD patients due to an increased risk of Torsades de Pointes in dialysis patients [205]. Similarly, flecainide toxicity is more severe in CKD patients [206]. Catheter ablation has shown reduced efficacy in CKD patients, with recurrence rates as high as 57%, especially in patients on dialysis [207, 208]. Cardioversion for sinus rhythm also has demonstrated lower success rates in CKD patients compared to those with normal kidney function [209]. Careful consideration should also be given to patients with impaired cardiac conduction system and reduced ejection fraction, as they do not tolerate class IC anti-arrhythmic drugs [210]. In fact, comorbidities should be considered in tandem with, if not prioritized over, kidney function when determining which therapies to withhold and which to prescribe for CKD-AF patients [211]. AF management in this population is not only focused on rate and rhythm control but also consider stroke prevention strategies. Anticoagulation therapy in CKD patients is particularly complex, as these patients are at an elevated risk of bleeding, complicating the safe use of anticoagulants like warfarin [212]. Unfortunately, limited clinical trials have specifically examined the effects of anticoagulation in CKD patients.
Some studies have shown a protective renal effect of estrogen administration in menopausal women with CKD [213]. However, dosage adjustment are necessary, as CKD women showed higher estrogen levels than those with normal renal function after receiving the same dose [214]. It is worth noting that estrogen replacement therapy has been associated with an increased risk of AF in the general population [215]. Manson et al. found that more than 60% of postmenopausal women treated with estrogen plus progestin developed new-onset AF, based on the Women’s Health Initiative study [216]. This proarrhythmic effect may be a consequence of estrogen’s modulation of RAAS and its ability to increase the expression of atrial natriuretic factor [217, 218]. Estrogen has also been shown to prolong APD and slow atrial conduction in murine atria, independently of the sex [219].
The effects of N-acetyl cysteine (NAC), a scavenger of free radicals and drug that binds to toxic metabolites, has been tested in CKD patients. Several studies showed that NAC did not provide protection against postoperative acute renal failure in patients with CKD undergoing cardiac surgery [220, 221]. On the other hand, other studies have shown that NAC can reduce the risk of CKD progression of any etiology to End-Stage Renal Disease (ESRD). For example, a Taiwanese cohort study of 123,000 CKD patients found that NAC use was associated with a reduced risk of progression to dialysis-dependent ESRD [222]. A meta-analysis of 15 trials with 768 CKD patients found that NAC improved eGFR and reduced serum creatinine compared to placebo, suggesting a benefit to kidney function. Additionally, NAC reduced cardiovascular events, which are closely linked to CKD progression [223]. Finally, a recent clinical trial (NCT04916080) has completed evaluating NAC in pediatric CKD patients, though results are pending.
Targeting inflammation is another emerging approach. A pilot, multicenter, randomized, placebo-controlled trial of the IL-1 receptor antagonist, anakinra, was tested in eight hemodialysis patients with CKD. The drug was well tolerated and did not increase infections or cytopenia. The median decrease in hsCRP over a 24-week study period was 41% in the anakinra group and 6% in the placebo group, which was not statistically significant due to the small sample size [224]. AstraZeneca recently launched a phase 1b clinical trial to assess the safety, tolerability, and pharmacodynamics of NLRP3 inhibitor AZD4144 in patients with established atherosclerotic cardiovascular and chronic kidney disease (NCT06675175). While AF is not a primary endpoint in these trials, post-hoc analyses may reveal effects on arrhythmia susceptibility. A separate feasibility trial (NCT00561093) examined the effects of a CKD-specific high-protein oral nutritional supplement with anti-inflammatory and antioxidant properties. Over 16 weeks, the results showed that this supplement was well tolerated and associated with a small but significant increase in serum albumin levels. However, the study was underpowered to determine effects on disease outcomes [225].
Finally, renal denervation (RDN) has emerged as a promising therapy. Combining RDN and with AF ablation has shown efficacy in reducing AF in CKD patients [226, 227]. Mechanistically, the beneficial effect of RDN on AF susceptibility, observed in a rodent CKD model induced by adenine-enriched diet, is attributed to the improve structural remodeling, reduced fibrosis, and enhanced electrical conduction within left atria of CKD rats [228]. Additionally, in a canine model of renal impairment, RDN also suppresses AF inducibility, which is associated with a significant decrease in pro-inflammatory NF-kB activation and IL-6 production [229]. Clinically, concurrent treatment utilizing both RDN and catheter ablation promote hemodynamic stability and reduces the risk of AF recurrence, especially in patients with hypertension [226, 227]. The efficacy of RDN on AF incidence depends on the type of AF, and the reduction of AF after RDN might be a secondary consequence of improved blood pressure or reduced sympathetic nervous system activity. Thus, renal denervation may be a good therapeutic treatment for preventing AF in CKD patients with concomitant resistant hypertension.
6. Perspectives
CKD constitutes a systemic risk factor for AF, promoting atrial arrhythmias in affected patients. Conversely, successful treatment of CKD may reduce the incidence of AF in certain patients suffering from both disorders. Nevertheless, studies have shown that even dialysis treatment may be insufficient to prevent the onset of AF in patients with advanced stages of CKD [230]. A deeper understanding of the molecular mechanisms underlying increased AF susceptibility in CKD patients could lead to the development and implementation of better treatment strategies. For example, an in-depth assessment of electrophysiological changes in atrial cardiomyocytes of CKD patients is currently lacking. Experimental models of CKD have shown prolonged APD in ventricular cardiomyocytes, attributed to increased late sodium current (INa,L) and decreased slowly inactivating potassium current (Ito,slow) [231]. The elevated INa,L can lead to intracellular Na+ overload, which subsequently enhances inward Ca2+ current through L-type Ca2+ channels. This prolongation may also be driven by the accumulation of uremic toxins due to impaired kidney function. Consistently, indoxyl sulfate, kynurenine, and kynurenic acid have been shown to prolong APD in hiPSC-derived cardiomyocytes by suppressing the rapid delayed rectifier potassium current (IKr) in a dose-dependent manner [232]. Additionally, increased cytoplasmic Ca2+ has been observed in pulmonary vein cardiomyocytes, resulting from enhanced PKA-mediated phosphorylation of RyR2 channels [3], supporting the role of triggered activity in AF development in CKD patients. Together, these findings support the notion that electrical remodeling and triggered activity are key proarrhythmic substrates induced by CKD. While electrophysiological studies in ventricular cardiomyocytes from CKD models may provide insights into potential AP changes in the context of CKD-associated AF, further research is needed to characterize the electrophysiological properties of atrial cardiomyocytes across different stages of CKD patients, with or without dialysis treatment. This line of investigation could help establish a foundation for research aimed at model validation and therapy development.
We discussed how multiple factors altered by kidney dysfunction, contribute to the development of cardiovascular disease, particularly AF. Given this connection, clinical studies are needed to evaluate whether preventing or treating kidney dysfunction can reduce the risk of AF. However, the etiology of kidney disease is highly diverse, and studies should be tailored accordingly. For example, the RAINIER study focuses on immunoglobulin A nephropathy by inhibiting B-cell activating factor (BAFF) and a proliferation-inducing ligand (APRIL), which are involved in B cell-mediated immune responses. Suppressing immune activity in this context may not only benefit kidney function but could also potentially lower the risk of AF. Notably, most CKD clinical trials to date have focused on showing CKD progression or reducing general cardiovascular events as outcome. AF remains an underexplored endpoint, a critical gap that should be addressed in future studies.
7. CONCLUSIONS
CKD is a pathology associated with various metabolic disorders that are significant risk factors for cardiovascular outcomes, such as AF. Structural and electrical atrial remodeling are key substrates for AF development. How CKD increases AF predisposition is not fully understood. A comprehensive review of systemic alterations commonly found in CKD patients showed that hyperglycemia, RAAS hyperactivations, mineral bone disorders, oxidative stress, and inflammation are all related to important AF substrates like triggered activity, atrial fibrosis, enlargement, and electrical remodeling. Whether these alterations exert a synergic or independent contribution to CKD-induced AF onset needs further investigation.
Acknowledgment:
This work was supposed by the National Institutes of Health [R01HL160992, R01HL147108, R01HL089598, R01HL153350, R01HL136389, R01HL164838, R01HL163277], and American Heart Association [936111].
Footnotes
Disclosure: None.
Declaration of Generative AI and AI-assisted technologies:
The authors did not use generative AI or AI-assisted technologies in the development of this manuscript.
REFERENCES
- [1].Kovesdy CP, Epidemiology of chronic kidney disease: an update 2022, Kidney Int Suppl (2011) 12 (2022) 7–11. 10.1016/j.kisu.2021.11.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [2].Chen TK, Knicely DH, Grams ME, Chronic Kidney Disease Diagnosis and Management: A Review, JAMA 322 (2019) 1294–1304. 10.1001/jama.2019.14745. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [3].Roth GA, Mensah GA, Johnson CO, Addolorato G, Ammirati E, Baddour LM, Barengo NC, Beaton AZ, Benjamin EJ, Benziger CP, Bonny A, Brauer M, Brodmann M, Cahill TJ, Carapetis J, Catapano AL, Chugh SS, Cooper LT, Coresh J, Criqui M, DeCleene N, Eagle KA, Emmons-Bell S, Feigin VL, Fernandez-Sola J, Fowkes G, Gakidou E, Grundy SM, He FJ, Howard G, Hu F, Inker L, Karthikeyan G, Kassebaum N, Koroshetz W, Lavie C, Lloyd-Jones D, Lu HS, Mirijello A, Temesgen AM, Mokdad A, Moran AE, Muntner P, Narula J, Neal B, Ntsekhe M, Moraes de Oliveira G, Otto C, Owolabi M, Pratt M, Rajagopalan S, Reitsma M, Ribeiro ALP, Rigotti N, Rodgers A, Sable C, Shakil S, Sliwa-Hahnle K, Stark B, Sundstrom J, Timpel P, Tleyjeh IM, Valgimigli M, Vos T, Whelton PK, Yacoub M, Zuhlke L, Murray C, Fuster V, G.-N.-J.G.B.o.C.D.W. Group, Global Burden of Cardiovascular Diseases and Risk Factors, 1990–2019: Update From the GBD 2019 Study, J Am Coll Cardiol 76 (2020) 2982–3021. 10.1016/j.jacc.2020.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Rovin BH, Adler SG, Barratt J, Bridoux F, Burdge KA, Chan TM, Cook HT, Fervenza FC, Gibson KL, Glassock RJ, Jayne DRW, Jha V, Liew A, Liu ZH, Mejia-Vilet JM, Nester CM, Radhakrishnan J, Rave EM, Reich HN, Ronco P, Sanders JF, Sethi S, Suzuki Y, Tang SCW, Tesar V, Vivarelli M, Wetzels JFM, Lytvyn L, Craig JC, Tunnicliffe DJ, Howell M, Tonelli MA, Cheung M, Earley A, Floege J, Executive summary of the KDIGO 2021 Guideline for the Management of Glomerular Diseases, Kidney Int 100 (2021) 753–779. 10.1016/j.kint.2021.05.015. [DOI] [PubMed] [Google Scholar]
- [5].Cruz DN, Gheorghiade M, Palazzuoli A, Ronco C, Bagshaw SM, Epidemiology and outcome of the cardio-renal syndrome, Heart Fail Rev 16 (2011) 531–42. 10.1007/s10741-010-9223-1. [DOI] [PubMed] [Google Scholar]
- [6].Schefold JC, Filippatos G, Hasenfuss G, Anker SD, von Haehling S, Heart failure and kidney dysfunction: epidemiology, mechanisms and management, Nat Rev Nephrol 12 (2016) 610–23. 10.1038/nrneph.2016.113. [DOI] [PubMed] [Google Scholar]
- [7].Ronco C, McCullough P, Anker SD, Anand I, Aspromonte N, Bagshaw SM, Bellomo R, Berl T, Bobek I, Cruz DN, Daliento L, Davenport A, Haapio M, Hillege H, House AA, Katz N, Maisel A, Mankad S, Zanco P, Mebazaa A, Palazzuoli A, Ronco F, Shaw A, Sheinfeld G, Soni S, Vescovo G, Zamperetti N, Ponikowski P, g. Acute Dialysis Quality Initiative consensus, Cardio-renal syndromes: report from the consensus conference of the acute dialysis quality initiative, Eur Heart J 31 (2010) 703–11. 10.1093/eurheartj/ehp507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Roy-Chaudhury P, Tumlin JA, Koplan BA, Costea AI, Kher V, Williamson D, Pokhariyal S, Charytan DM, Mi D.i., committees, Primary outcomes of the Monitoring in Dialysis Study indicate that clinically significant arrhythmias are common in hemodialysis patients and related to dialytic cycle, Kidney Int 93 (2018) 941–951. 10.1016/j.kint.2017.11.019. [DOI] [PubMed] [Google Scholar]
- [9].Charytan DM, Foley R, McCullough PA, Rogers JD, Zimetbaum P, Herzog CA, Tumlin JA, Mi DI, Committees, Arrhythmia and Sudden Death in Hemodialysis Patients: Protocol and Baseline Characteristics of the Monitoring in Dialysis Study, Clin J Am Soc Nephrol 11 (2016) 721–34. 10.2215/CJN.09350915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Akoum N, Zelnick LR, de Boer IH, Hirsch IB, Trence D, Henry C, Robinson N, Bansal N, Rates of Cardiac Rhythm Abnormalities in Patients with CKD and Diabetes, Clin J Am Soc Nephrol 14 (2019) 549–556. 10.2215/CJN.09420818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Soliman EZ, Prineas RJ, Go AS, Xie D, Lash JP, Rahman M, Ojo A, Teal VL, Jensvold NG, Robinson NL, Dries DL, Bazzano L, Mohler ER, Wright JT, Feldman HI, Chronic G Renal Insufficiency Cohort Study, Chronic kidney disease and prevalent atrial fibrillation: the Chronic Renal Insufficiency Cohort (CRIC), Am Heart J 159 (2010) 1102–7. 10.1016/j.ahj.2010.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Hindricks G, Potpara T, Dagres N, Arbelo E, Bax JJ, Blomstrom-Lundqvist C, Boriani G, Castella M, Dan GA, Dilaveris PE, Fauchier L, Filippatos G, Kalman JM, La Meir M, Lane DA, Lebeau JP, Lettino M, Lip GYH, Pinto FJ, Thomas GN, Valgimigli M, Van Gelder IC, Van Putte BP, Watkins CL, E.S.C.S.D. Group, 2020 ESC Guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): The Task Force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) Developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC, Eur Heart J 42 (2021) 373–498. 10.1093/eurheartj/ehaa612. [DOI] [PubMed] [Google Scholar]
- [13].Heijman J, Algalarrondo V, Voigt N, Melka J, Wehrens XH, Dobrev D, Nattel S, The value of basic research insights into atrial fibrillation mechanisms as a guide to therapeutic innovation: a critical analysis, Cardiovasc Res 109 (2016) 467–79. 10.1093/cvr/cvv275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Nattel S, Heijman J, Zhou L, Dobrev D, Molecular Basis of Atrial Fibrillation Pathophysiology and Therapy: A Translational Perspective, Circ Res 127 (2020) 51–72. 10.1161/CIRCRESAHA.120.316363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Ananthapanyasut W, Napan S, Rudolph EH, Harindhanavudhi T, Ayash H, Guglielmi KE, Lerma EV, Prevalence of atrial fibrillation and its predictors in nondialysis patients with chronic kidney disease, Clin J Am Soc Nephrol 5 (2010) 173–81. 10.2215/CJN.03170509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Wetmore JB, Mahnken JD, Rigler SK, Ellerbeck EF, Mukhopadhyay P, Spertus JA, Hou Q, Shireman TI, The prevalence of and factors associated with chronic atrial fibrillation in Medicare/Medicaid-eligible dialysis patients, Kidney Int 81 (2012) 469–76. 10.1038/ki.2011.416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Watanabe H, Watanabe T, Sasaki S, Nagai K, Roden DM, Aizawa Y, Close bidirectional relationship between chronic kidney disease and atrial fibrillation: the Niigata preventive medicine study, Am Heart J 158 (2009) 629–36. 10.1016/j.ahj.2009.06.031. [DOI] [PubMed] [Google Scholar]
- [18].Ha JT, Freedman SB, Kelly DM, Neuen BL, Perkovic V, Jun M, Badve SV, Kidney Function, Albuminuria, and Risk of Incident Atrial Fibrillation: A Systematic Review and Meta-Analysis, Am J Kidney Dis 83 (2024) 350–359 e1. 10.1053/j.ajkd.2023.07.023. [DOI] [PubMed] [Google Scholar]
- [19].Bansal N, Xie D, Sha D, Appel LJ, Deo R, Feldman HI, He J, Jamerson K, Kusek JW, Messe S, Navaneethan SD, Rahman M, Ricardo AC, Soliman EZ, Townsend R, Go AS, Cardiovascular Events after New-Onset Atrial Fibrillation in Adults with CKD: Results from the Chronic Renal Insufficiency Cohort (CRIC) Study, J Am Soc Nephrol 29 (2018) 2859–2869. 10.1681/ASN.2018050514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Go AS, Fang MC, Udaltsova N, Chang Y, Pomernacki NK, Borowsky L, Singer DE, Investigators AS, Impact of proteinuria and glomerular filtration rate on risk of thromboembolism in atrial fibrillation: the anticoagulation and risk factors in atrial fibrillation (ATRIA) study, Circulation 119 (2009) 1363–9. 10.1161/CIRCULATIONAHA.108.816082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Massicotte-Azarniouch D, Kuwornu JP, Carrero JJ, Lam NN, Molnar AO, Zimmerman D, McCallum MK, Garg AX, Sood MM, Incident Atrial Fibrillation and the Risk of Congestive Heart Failure, Myocardial Infarction, End-Stage Kidney Disease, and Mortality Among Patients With a Decreased Estimated GFR, Am J Kidney Dis 71 (2018) 191–199. 10.1053/j.ajkd.2017.08.016. [DOI] [PubMed] [Google Scholar]
- [22].Kumar S, Lim E, Covic A, Verhamme P, Gale CP, Camm AJ, Goldsmith D, Anticoagulation in Concomitant Chronic Kidney Disease and Atrial Fibrillation: JACC Review Topic of the Week, J Am Coll Cardiol 74 (2019) 2204–2215. 10.1016/j.jacc.2019.08.1031. [DOI] [PubMed] [Google Scholar]
- [23].Horio T, Iwashima Y, Kamide K, Tokudome T, Yoshihara F, Nakamura S, Kawano Y, Chronic kidney disease as an independent risk factor for new-onset atrial fibrillation in hypertensive patients, J Hypertens 28 (2010) 1738–44. 10.1097/HJH.0b013e32833a7dfe. [DOI] [PubMed] [Google Scholar]
- [24].Shang W, Li L, Huang S, Zeng R, Huang L, Ge S, Xu G, Chronic Kidney Disease and the Risk of New-Onset Atrial Fibrillation: A Meta-Analysis of Prospective Cohort Studies, PLoS One 11 (2016) e0155581. 10.1371/journal.pone.0155581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Ansari N, Manis T, Feinfeld DA, Symptomatic atrial arrhythmias in hemodialysis patients, Ren Fail 23 (2001) 71–6. 10.1081/jdi-100001285. [DOI] [PubMed] [Google Scholar]
- [26].Korantzopoulos P, Kolettis TM, Galaris D, Goudevenos JA, The role of oxidative stress in the pathogenesis and perpetuation of atrial fibrillation, Int J Cardiol 115 (2007) 135–43. 10.1016/j.ijcard.2006.04.026. [DOI] [PubMed] [Google Scholar]
- [27].Turakhia MP, Blankestijn PJ, Carrero JJ, Clase CM, Deo R, Herzog CA, Kasner SE, Passman RS, Pecoits-Filho R, Reinecke H, Shroff GR, Zareba W, Cheung M, Wheeler DC, Winkelmayer WC, Wanner C, Conference P, Chronic kidney disease and arrhythmias: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference, Eur Heart J 39 (2018) 2314–2325. 10.1093/eurheartj/ehy060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Baber U, Howard VJ, Halperin JL, Soliman EZ, Zhang X, McClellan W, Warnock DG, Muntner P, Association of chronic kidney disease with atrial fibrillation among adults in the United States: REasons for Geographic and Racial Differences in Stroke (REGARDS) Study, Circ Arrhythm Electrophysiol 4 (2011) 26–32. 10.1161/CIRCEP.110.957100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Ahmed SB, Ramesh S, Sex hormones in women with kidney disease, Nephrol Dial Transplant 31 (2016) 1787–1795. 10.1093/ndt/gfw084. [DOI] [PubMed] [Google Scholar]
- [30].Zitzmann M, Testosterone deficiency and chronic kidney disease, J Clin Transl Endocrinol 37 (2024) 100365. 10.1016/j.jcte.2024.100365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Cevik EC, Erel CT, Ozcivit Erkan IB, Sarafidis P, Armeni E, Fistonic I, Hillard T, Hirschberg AL, Meczekalski B, Mendoza N, Mueck AO, Simoncini T, Stute P, van Dijken D, Rees M, Lambrinoudaki I, Chronic kidney disease and menopausal health: An EMAS clinical guide, Maturitas 192 (2025) 108145. 10.1016/j.maturitas.2024.108145. [DOI] [PubMed] [Google Scholar]
- [32].Hatamizadeh P, Fonarow GC, Budoff MJ, Darabian S, Kovesdy CP, Kalantar-Zadeh K, Cardiorenal syndrome: pathophysiology and potential targets for clinical management, Nat Rev Nephrol 9 (2013) 99–111. 10.1038/nrneph.2012.279. [DOI] [PubMed] [Google Scholar]
- [33].Zhao Y, Malik S, Budoff MJ, Correa A, Ashley KE, Selvin E, Watson KE, Wong ND, Identification and Predictors for Cardiovascular Disease Risk Equivalents among Adults With Diabetes Mellitus, Diabetes Care (2021). 10.2337/dc21-0431. [DOI] [PubMed] [Google Scholar]
- [34].Heo NJ, Rhee SY, Waalen J, Steinhubl S, Chronic kidney disease and undiagnosed atrial fibrillation in individuals with diabetes, Cardiovasc Diabetol 19 (2020) 157. 10.1186/s12933-020-01128-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Escobar Vasco MA, Fantaye SH, Raghunathan S, Solis-Herrera C, The potential role of finerenone in patients with type 1 diabetes and chronic kidney disease, Diabetes Obes Metab 26 (2024) 4135–4146. 10.1111/dom.15773. [DOI] [PubMed] [Google Scholar]
- [36].Kumar M, Dev S, Khalid MU, Siddenthi SM, Noman M, John C, Akubuiro C, Haider A, Rani R, Kashif M, Varrassi G, Khatri M, Kumar S, Mohamad T, The Bidirectional Link Between Diabetes and Kidney Disease: Mechanisms and Management, Cureus 15 (2023) e45615. 10.7759/cureus.45615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Yosefy O, Sharon B, Yagil C, Shlapoberski M, Livoff A, Novitski I, Beeri R, Yagil Y, Yosefy C, Diabetes induces remodeling of the left atrial appendage independently of atrial fibrillation in a rodent model of type-2 diabetes, Cardiovasc Diabetol 20 (2021) 149. 10.1186/s12933-021-01347-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Dyntar D, Sergeev P, Klisic J, Ambuhl P, Schaub MC, Donath MY, High glucose alters cardiomyocyte contacts and inhibits myofibrillar formation, J Clin Endocrinol Metab 91 (2006) 1961–7. 10.1210/jc.2005-1904. [DOI] [PubMed] [Google Scholar]
- [39].Pandey S, Madreiter-Sokolowski CT, Mangmool S, Parichatikanond W, High Glucose-Induced Cardiomyocyte Damage Involves Interplay between Endothelin ET-1/ET(A)/ET(B) Receptor and mTOR Pathway, Int J Mol Sci 23 (2022). 10.3390/ijms232213816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Smogorzewski M, Galfayan V, Massry SG, High glucose concentration causes a rise in [Ca2+]i of cardiac myocytes, Kidney Int 53 (1998) 1237–43. 10.1046/j.1523-1755.1998.00868.x. [DOI] [PubMed] [Google Scholar]
- [41].Komada T, Muruve DA, The role of inflammasomes in kidney disease, Nat Rev Nephrol 15 (2019) 501–520. 10.1038/s41581-019-0158-z. [DOI] [PubMed] [Google Scholar]
- [42].Nagata D, Hishida E, Elucidating the complex interplay between chronic kidney disease and hypertension, Hypertens Res (2024). 10.1038/s41440-024-01937-8. [DOI] [PubMed] [Google Scholar]
- [43].Lau YF, Yiu KH, Siu CW, Tse HF, Hypertension and atrial fibrillation: epidemiology, pathophysiology and therapeutic implications, Journal of Human Hypertension 26 (2012) 563–569. 10.1038/jhh.2011.105. [DOI] [PubMed] [Google Scholar]
- [44].Gawałko M, Linz D, Atrial Fibrillation Detection and Management in Hypertension, Hypertension 80 (2023) 523–533. 10.1161/hypertensionaha.122.19459. [DOI] [PubMed] [Google Scholar]
- [45].Goette A, Staack T, Rocken C, Arndt M, Geller JC, Huth C, Ansorge S, Klein HU, Lendeckel U, Increased expression of extracellular signal-regulated kinase and angiotensin-converting enzyme in human atria during atrial fibrillation, J Am Coll Cardiol 35 (2000) 1669–77. 10.1016/s0735-1097(00)00611-2. [DOI] [PubMed] [Google Scholar]
- [46].Milliez P, Deangelis N, Rucker-Martin C, Leenhardt A, Vicaut E, Robidel E, Beaufils P, Delcayre C, Hatem SN, Spironolactone reduces fibrosis of dilated atria during heart failure in rats with myocardial infarction, Eur Heart J 26 (2005) 2193–9. 10.1093/eurheartj/ehi478. [DOI] [PubMed] [Google Scholar]
- [47].Goette A, Arndt M, Rocken C, Spiess A, Staack T, Geller JC, Huth C, Ansorge S, Klein HU, Lendeckel U, Regulation of angiotensin II receptor subtypes during atrial fibrillation in humans, Circulation 101 (2000) 2678–81. 10.1161/01.cir.101.23.2678. [DOI] [PubMed] [Google Scholar]
- [48].Xiao HD, Fuchs S, Campbell DJ, Lewis W, Dudley SC Jr., Kasi VS, Hoit BD, Keshelava G, Zhao H, Capecchi MR, Bernstein KE, Mice with cardiac-restricted angiotensin-converting enzyme (ACE) have atrial enlargement, cardiac arrhythmia, and sudden death, Am J Pathol 165 (2004) 1019–32. 10.1016/S0002-9440(10)63363-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Yin Y, Dalal D, Liu Z, Wu J, Liu D, Lan X, Dai Y, Su L, Ling Z, She Q, Luo K, Woo K, Dong J, Prospective randomized study comparing amiodarone vs. amiodarone plus losartan vs. amiodarone plus perindopril for the prevention of atrial fibrillation recurrence in patients with lone paroxysmal atrial fibrillation, Eur Heart J 27 (2006) 1841–6. 10.1093/eurheartj/ehl135. [DOI] [PubMed] [Google Scholar]
- [50].Liao J, Zhang S, Yang S, Lu Y, Lu K, Wu Y, Wu Q, Zhao N, Dong Q, Chen L, Du Y, Interleukin-6-Mediated-Ca(2+) Handling Abnormalities Contributes to Atrial Fibrillation in Sterile Pericarditis Rats, Front Immunol 12 (2021) 758157. 10.3389/fimmu.2021.758157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Purohit A, Rokita AG, Guan X, Chen B, Koval OM, Voigt N, Neef S, Sowa T, Gao Z, Luczak ED, Stefansdottir H, Behunin AC, Li N, El-Accaoui RN, Yang B, Swaminathan PD, Weiss RM, Wehrens XH, Song LS, Dobrev D, Maier LS, Anderson ME, Oxidized Ca(2+)/calmodulin-dependent protein kinase II triggers atrial fibrillation, Circulation 128 (2013) 1748–57. 10.1161/CIRCULATIONAHA.113.003313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Zhang M, Prosser BL, Bamboye MA, Gondim ANS, Santos CX, Martin D, Ghigo A, Perino A, Brewer AC, Ward CW, Hirsch E, Lederer WJ, Shah AM, Contractile Function During Angiotensin-II Activation: Increased Nox2 Activity Modulates Cardiac Calcium Handling via Phospholamban Phosphorylation, J Am Coll Cardiol 66 (2015) 261–272. 10.1016/j.jacc.2015.05.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Liu Y, Lv H, Tan R, An X, Niu XH, Liu YJ, Yang X, Yin X, Xia YL, Platelets Promote Ang II (Angiotensin II)-Induced Atrial Fibrillation by Releasing TGF-beta1 (Transforming Growth Factor-beta1) and Interacting With Fibroblasts, Hypertension 76 (2020) 1856–1867. 10.1161/HYPERTENSIONAHA.120.15016. [DOI] [PubMed] [Google Scholar]
- [54].Matsushita N, Ishida N, Ibi M, Saito M, Takahashi M, Taniguchi S, Iwakura Y, Morino Y, Taira E, Sawa Y, Hirose M, IL-1β Plays an Important Role in Pressure Overload-Induced Atrial Fibrillation in Mice, Biological and Pharmaceutical Bulletin 42 (2019) 543–546. 10.1248/bpb.b18-00363. [DOI] [PubMed] [Google Scholar]
- [55].Moreira LM, Takawale A, Hulsurkar M, Menassa DA, Antanaviciute A, Lahiri SK, Mehta N, Evans N, Psarros C, Robinson P, Sparrow AJ, Gillis MA, Ashley N, Naud P, Barallobre-Barreiro J, Theofilatos K, Lee A, Norris M, Clarke MV, Russell PK, Casadei B, Bhattacharya S, Zajac JD, Davey RA, Sirois M, Mead A, Simmons A, Mayr M, Sayeed R, Krasopoulos G, Redwood C, Channon KM, Tardif JC, Wehrens XHT, Nattel S, Reilly S, Paracrine signalling by cardiac calcitonin controls atrial fibrogenesis and arrhythmia, Nature 587 (2020) 460–465. 10.1038/s41586-020-2890-8. [DOI] [PubMed] [Google Scholar]
- [56].Rouached M, El Kadiri Boutchich S, Al Rifai AM, Garabedian M, Fournier A, Prevalence of abnormal serum vitamin D, PTH, calcium, and phosphorus in patients with chronic kidney disease: results of the study to evaluate early kidney disease, Kidney Int 74 (2008) 389–90. 10.1038/ki.2008.169. [DOI] [PubMed] [Google Scholar]
- [57].Wang Q, Su W, Shen Z, Wang R, Correlation between Soluble alpha-Klotho and Renal Function in Patients with Chronic Kidney Disease: A Review and Meta-Analysis, Biomed Res Int 2018 (2018) 9481475. 10.1155/2018/9481475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Henaut L, Chillon JM, Kamel S, Massy ZA, Updates on the Mechanisms and the Care of Cardiovascular Calcification in Chronic Kidney Disease, Semin Nephrol 38 (2018) 233–250. 10.1016/j.semnephrol.2018.02.004. [DOI] [PubMed] [Google Scholar]
- [59].Viegas C, Araujo N, Marreiros C, Simes D, The interplay between mineral metabolism, vascular calcification and inflammation in Chronic Kidney Disease (CKD): challenging old concepts with new facts, Aging (Albany NY) 11 (2019) 4274–4299. 10.18632/aging.102046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Mehta R, Cai X, Lee J, Scialla JJ, Bansal N, Sondheimer JH, Chen J, Hamm LL, Ricardo AC, Navaneethan SD, Deo R, Rahman M, Feldman HI, Go AS, Isakova T, Wolf M, Chronic I Renal Insufficiency Cohort Study, Association of Fibroblast Growth Factor 23 With Atrial Fibrillation in Chronic Kidney Disease, From the Chronic Renal Insufficiency Cohort Study, JAMA Cardiol 1 (2016) 548–56. 10.1001/jamacardio.2016.1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Taniguchi M, Fukagawa M, Fujii N, Hamano T, Shoji T, Yokoyama K, Nakai S, Shigematsu T, Iseki K, Tsubakihara Y, T. Committee of Renal Data Registry of the Japanese Society for Dialysis, Serum phosphate and calcium should be primarily and consistently controlled in prevalent hemodialysis patients, Ther Apher Dial 17 (2013) 221–8. 10.1111/1744-9987.12030. [DOI] [PubMed] [Google Scholar]
- [62].Lee SH, Lee MY, Shin SY, Lee WS, Kim SW, Park SJ, Kim JS, Sung KC, Relationship between Cardiovascular Calcium and Atrial Fibrillation, J Clin Med 11 (2022). 10.3390/jcm11020371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Lopez FL, Agarwal SK, Grams ME, Loehr LR, Soliman EZ, Lutsey PL, Chen LY, Huxley RR, Alonso A, Relation of serum phosphorus levels to the incidence of atrial fibrillation (from the Atherosclerosis Risk In Communities [ARIC] study), Am J Cardiol 111 (2013) 857–62. 10.1016/j.amjcard.2012.11.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Wu Y, Kong XJ, Ji YY, Fan J, Ji CC, Chen XM, Ma YD, Tang AL, Cheng YJ, Wu SH, Serum electrolyte concentrations and risk of atrial fibrillation: an observational and mendelian randomization study, BMC Genomics 25 (2024) 280. 10.1186/s12864-024-10197-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Hsu YJ, Chang GJ, Lai YJ, Chan YH, Chen WJ, Kuo CT, Yeh YH, High-phosphate diet causes atrial remodeling and increases atrial fibrillation vulnerability via STAT3/NF-kappaB signaling and oxidative stress, Acta Physiol (Oxf) 238 (2023) e13964. 10.1111/apha.13964. [DOI] [PubMed] [Google Scholar]
- [66].Voigt N, Li N, Wang Q, Wang W, Trafford AW, Abu-Taha I, Sun Q, Wieland T, Ravens U, Nattel S, Wehrens XH, Dobrev D, Enhanced sarcoplasmic reticulum Ca2+ leak and increased Na+-Ca2+ exchanger function underlie delayed afterdepolarizations in patients with chronic atrial fibrillation, Circulation 125 (2012) 2059–70. 10.1161/CIRCULATIONAHA.111.067306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Holden RM, Norman PA, Day AG, Silver SA, Clemens KK, Iliescu E, Vitamin D Status and Treatment in ESKD: Links to Improved CKD-MBD Laboratory Parameters in a Real-World Setting, Am J Nephrol (2024) 1–9. 10.1159/000541109. [DOI] [PubMed] [Google Scholar]
- [68].Nigwekar SU, Tamez H, Thadhani RI, Vitamin D and chronic kidney disease-mineral bone disease (CKD-MBD), Bonekey Rep 3 (2014) 498. 10.1038/bonekey.2013.232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Li L, Zhao J, Association of serum 25-hydroxyvitamin D with cardiovascular and all-cause mortality in patients with chronic kidney disease: NHANES 2007‒2018 results, Clinics (Sao Paulo) 79 (2024) 100437. 10.1016/j.clinsp.2024.100437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [70].Liu X, Wang W, Tan Z, Zhu X, Liu M, Wan R, Hong K, The relationship between vitamin D and risk of atrial fibrillation: a dose-response analysis of observational studies, Nutr J 18 (2019) 73. 10.1186/s12937-019-0485-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Virtanen JK, Hantunen S, Lamberg-Allardt C, Manson JE, Nurmi T, Uusitupa M, Voutilainen A, Tuomainen TP, The effect of vitamin D(3) supplementation on atrial fibrillation in generally healthy men and women: The Finnish Vitamin D Trial, Am Heart J 264 (2023) 177–182. 10.1016/j.ahj.2023.05.024. [DOI] [PubMed] [Google Scholar]
- [72].Fusaro M, Gallieni M, Rebora P, Rizzo MA, Luise MC, Riva H, Bertoli S, Conte F, Stella A, Ondei P, Rossi E, Valsecchi MG, Santoro A, Genovesi S, Atrial fibrillation and low vitamin D levels are associated with severe vascular calcifications in hemodialysis patients, J Nephrol 29 (2016) 419–426. 10.1007/s40620-015-0236-7. [DOI] [PubMed] [Google Scholar]
- [73].Chen S, Glenn DJ, Ni W, Grigsby CL, Olsen K, Nishimoto M, Law CS, Gardner DG, Expression of the vitamin d receptor is increased in the hypertrophic heart, Hypertension 52 (2008) 1106–12. 10.1161/HYPERTENSIONAHA.108.119602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Rahman A, Hershey S, Ahmed S, Nibbelink K, Simpson RU, Heart extracellular matrix gene expression profile in the vitamin D receptor knockout mice, J Steroid Biochem Mol Biol 103 (2007) 416–9. 10.1016/j.jsbmb.2006.12.081. [DOI] [PubMed] [Google Scholar]
- [75].Koleganova N, Piecha G, Ritz E, Gross ML, Calcitriol ameliorates capillary deficit and fibrosis of the heart in subtotally nephrectomized rats, Nephrol Dial Transplant 24 (2009) 778–87. 10.1093/ndt/gfn549. [DOI] [PubMed] [Google Scholar]
- [76].Mizobuchi M, Nakamura H, Tokumoto M, Finch J, Morrissey J, Liapis H, Slatopolsky E, Myocardial effects of VDR activators in renal failure, J Steroid Biochem Mol Biol 121 (2010) 188–92. 10.1016/j.jsbmb.2010.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Yu W, Dong X, Dan G, Ye F, Cheng J, Zhao Y, Chen M, Sai Y, Zou Z, Vitamin D3 protects against nitrogen mustard-induced apoptosis of the bronchial epithelial cells via activating the VDR/Nrf2/Sirt3 pathway, Toxicol Lett 354 (2022) 14–23. 10.1016/j.toxlet.2021.10.016. [DOI] [PubMed] [Google Scholar]
- [78].F. National Kidney, K/DOQI clinical practice guidelines for bone metabolism and disease in chronic kidney disease, Am J Kidney Dis 42 (2003) S1–201. [PubMed] [Google Scholar]
- [79].Folsom AR, Alonso A, Misialek JR, Michos ED, Selvin E, Eckfeldt JH, Coresh J, Pankow JS, Lutsey PL, Parathyroid hormone concentration and risk of cardiovascular diseases: the Atherosclerosis Risk in Communities (ARIC) study, Am Heart J 168 (2014) 296–302. 10.1016/j.ahj.2014.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Platt A, Wilson J, Hall R, Ephraim PL, Morton S, Shafi T, Weiner DE, Boulware LE, Pendergast J, Scialla JJ, Comparative G Effectiveness Studies in Dialysis Patients, Comparative Effectiveness of Alternative Treatment Approaches to Secondary Hyperparathyroidism in Patients Receiving Maintenance Hemodialysis: An Observational Trial Emulation, Am J Kidney Dis 83 (2024) 58–70. 10.1053/j.ajkd.2023.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Rienstra M, Lubitz SA, Zhang ML, Cooper RR, Ellinor PT, Elevation of parathyroid hormone levels in atrial fibrillation, J Am Coll Cardiol 57 (2011) 2542–3. 10.1016/j.jacc.2011.01.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Lee KH, Shin MH, Park HW, Cho JG, Kweon SS, Lee YH, Association between Serum Parathyroid Hormone Levels and the Prevalence of Atrial Fibrillation: the Dong-gu Study, Korean Circ J 48 (2018) 159–167. 10.4070/kcj.2017.0187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Smogorzewski M, Zayed M, Zhang YB, Roe J, Massry SG, Parathyroid hormone increases cytosolic calcium concentration in adult rat cardiac myocytes, Am J Physiol 264 (1993) H1998–2006. 10.1152/ajpheart.1993.264.6.H1998. [DOI] [PubMed] [Google Scholar]
- [84].Wehrens XH, Lehnart SE, Huang F, Vest JA, Reiken SR, Mohler PJ, Sun J, Guatimosim S, Song LS, Rosemblit N, D’Armiento JM, Napolitano C, Memmi M, Priori SG, Lederer WJ, Marks AR, FKBP12.6 deficiency and defective calcium release channel (ryanodine receptor) function linked to exercise-induced sudden cardiac death, Cell 113 (2003) 829–40. 10.1016/s0092-8674(03)00434-3. [DOI] [PubMed] [Google Scholar]
- [85].Tastan I, Schreckenberg R, Mufti S, Abdallah Y, Piper HM, Schluter KD, Parathyroid hormone improves contractile performance of adult rat ventricular cardiomyocytes at low concentrations in a non-acute way, Cardiovasc Res 82 (2009) 77–83. 10.1093/cvr/cvp027. [DOI] [PubMed] [Google Scholar]
- [86].Navarro-Garcia JA, Gonzalez-Lafuente L, Fernandez-Velasco M, Ruilope LM, Ruiz-Hurtado G, Fibroblast Growth Factor-23-Klotho Axis in Cardiorenal Syndrome: Mediators and Potential Therapeutic Targets, Front Physiol 12 (2021) 775029. 10.3389/fphys.2021.775029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [87].Touchberry CD, Green TM, Tchikrizov V, Mannix JE, Mao TF, Carney BW, Girgis M, Vincent RJ, Wetmore LA, Dawn B, Bonewald LF, Stubbs JR, Wacker MJ, FGF23 is a novel regulator of intracellular calcium and cardiac contractility in addition to cardiac hypertrophy, Am J Physiol Endocrinol Metab 304 (2013) E863–73. 10.1152/ajpendo.00596.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Faul C, Amaral AP, Oskouei B, Hu MC, Sloan A, Isakova T, Gutierrez OM, Aguillon-Prada R, Lincoln J, Hare JM, Mundel P, Morales A, Scialla J, Fischer M, Soliman EZ, Chen J, Go AS, Rosas SE, Nessel L, Townsend RR, Feldman HI, St John Sutton M, Ojo A, Gadegbeku C, Di Marco GS, Reuter S, Kentrup D, Tiemann K, Brand M, Hill JA, Moe OW, Kuro OM, Kusek JW, Keane MG, Wolf M, FGF23 induces left ventricular hypertrophy, J Clin Invest 121 (2011) 4393–408. 10.1172/JCI46122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Grabner A, Schramm K, Silswal N, Hendrix M, Yanucil C, Czaya B, Singh S, Wolf M, Hermann S, Stypmann J, Di Marco GS, Brand M, Wacker MJ, Faul C, FGF23/FGFR4-mediated left ventricular hypertrophy is reversible, Sci Rep 7 (2017) 1993. 10.1038/s41598-017-02068-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Mathew JS, Sachs MC, Katz R, Patton KK, Heckbert SR, Hoofnagle AN, Alonso A, Chonchol M, Deo R, Ix JH, Siscovick DS, Kestenbaum B, de Boer IH, Fibroblast growth factor-23 and incident atrial fibrillation: the Multi-Ethnic Study of Atherosclerosis (MESA) and the Cardiovascular Health Study (CHS), Circulation 130 (2014) 298–307. 10.1161/CIRCULATIONAHA.113.005499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Navarro-Garcia JA, Delgado C, Fernandez-Velasco M, Val-Blasco A, Rodriguez-Sanchez E, Aceves-Ripoll J, Gomez-Hurtado N, Bada-Bosch T, Merida-Herrero E, Hernandez E, Praga M, Salguero R, Solis J, Arribas F, Delgado JF, Bueno H, Kuro OM, Ruilope LM, Ruiz-Hurtado G, Fibroblast growth factor-23 promotes rhythm alterations and contractile dysfunction in adult ventricular cardiomyocytes, Nephrol Dial Transplant 34 (2019) 1864–1875. 10.1093/ndt/gfy392. [DOI] [PubMed] [Google Scholar]
- [92].Pastor-Arroyo EM, Gehring N, Krudewig C, Costantino S, Bettoni C, Knopfel T, Sabrautzki S, Lorenz-Depiereux B, Pastor J, Strom TM, Hrabe de Angelis M, Camici GG, Paneni F, Wagner CA, Rubio-Aliaga I, The elevation of circulating fibroblast growth factor 23 without kidney disease does not increase cardiovascular disease risk, Kidney Int 94 (2018) 49–59. 10.1016/j.kint.2018.02.017. [DOI] [PubMed] [Google Scholar]
- [93].Nowak A, Friedrich B, Artunc F, Serra AL, Breidthardt T, Twerenbold R, Peter M, Mueller C, Prognostic value and link to atrial fibrillation of soluble Klotho and FGF23 in hemodialysis patients, PLoS One 9 (2014) e100688. 10.1371/journal.pone.0100688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Mizia-Stec K, Wieczorek J, Polak M, Wybraniec MT, Wozniak-Skowerska I, Hoffmann A, Nowak S, Wikarek M, Wnuk-Wojnar A, Chudek J, Wiecek A, Lower soluble Klotho and higher fibroblast growth factor 23 serum levels are associated with episodes of atrial fibrillation, Cytokine 111 (2018) 106–111. 10.1016/j.cyto.2018.08.005. [DOI] [PubMed] [Google Scholar]
- [95].Hung Y, Chung CC, Chen YC, Kao YH, Lin WS, Chen SA, Chen YJ, Klotho Modulates Pro-Fibrotic Activities in Human Atrial Fibroblasts through Inhibition of Phospholipase C Signaling and Suppression of Store-Operated Calcium Entry, Biomedicines 10 (2022). 10.3390/biomedicines10071574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Ruiz S, Pergola PE, Zager RA, Vaziri ND, Targeting the transcription factor Nrf2 to ameliorate oxidative stress and inflammation in chronic kidney disease, Kidney Int 83 (2013) 1029–41. 10.1038/ki.2012.439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Himmelfarb J, Stenvinkel P, Ikizler TA, Hakim RM, The elephant in uremia: oxidant stress as a unifying concept of cardiovascular disease in uremia, Kidney Int 62 (2002) 1524–38. 10.1046/j.1523-1755.2002.00600.x. [DOI] [PubMed] [Google Scholar]
- [98].Drozdz D, Kwinta P, Sztefko K, Kordon Z, Drozdz T, Latka M, Miklaszewska M, Zachwieja K, Rudzinski A, Pietrzyk JA, Oxidative Stress Biomarkers and Left Ventricular Hypertrophy in Children with Chronic Kidney Disease, Oxid Med Cell Longev 2016 (2016) 7520231. 10.1155/2016/7520231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].Gondouin B, Jourde-Chiche N, Sallee M, Dou L, Cerini C, Loundou A, Morange S, Berland Y, Burtey S, Brunet P, Guieu R, Dussol B, Plasma Xanthine Oxidase Activity Is Predictive of Cardiovascular Disease in Patients with Chronic Kidney Disease, Independently of Uric Acid Levels, Nephron 131 (2015) 167–74. 10.1159/000441091. [DOI] [PubMed] [Google Scholar]
- [100].Niwa T, Indoxyl sulfate is a nephro-vascular toxin, J Ren Nutr 20 (2010) S2–6. 10.1053/j.jrn.2010.05.002. [DOI] [PubMed] [Google Scholar]
- [101].Garcia-Bello JA, Gomez-Diaz RA, Contreras-Rodriguez A, Talavera JO, Mondragon-Gonzalez R, Sanchez-Barbosa L, Diaz-Flores M, Valladares-Salgado A, Gallardo JM, Aguilar-Kitsu A, Lagunas-Munoz J, Wacher NH, Carotid intima media thickness, oxidative stress, and inflammation in children with chronic kidney disease, Pediatr Nephrol 29 (2014) 273–81. 10.1007/s00467-013-2626-1. [DOI] [PubMed] [Google Scholar]
- [102].Luczak ED, Anderson ME, CaMKII oxidative activation and the pathogenesis of cardiac disease, J Mol Cell Cardiol 73 (2014) 112–6. 10.1016/j.yjmcc.2014.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Erickson JR, Joiner ML, Guan X, Kutschke W, Yang J, Oddis CV, Bartlett RK, Lowe JS, O’Donnell SE, Aykin-Burns N, Zimmerman MC, Zimmerman K, Ham AJ, Weiss RM, Spitz DR, Shea MA, Colbran RJ, Mohler PJ, Anderson ME, A dynamic pathway for calcium-independent activation of CaMKII by methionine oxidation, Cell 133 (2008) 462–74. 10.1016/j.cell.2008.02.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Wagner S, Ruff HM, Weber SL, Bellmann S, Sowa T, Schulte T, Anderson ME, Grandi E, Bers DM, Backs J, Belardinelli L, Maier LS, Reactive oxygen species-activated Ca/calmodulin kinase IIdelta is required for late I(Na) augmentation leading to cellular Na and Ca overload, Circ Res 108 (2011) 555–65. 10.1161/CIRCRESAHA.110.221911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Pfenniger A, Yoo S, Arora R, Oxidative stress and atrial fibrillation, J Mol Cell Cardiol 196 (2024) 141–151. 10.1016/j.yjmcc.2024.09.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Akchurin OM, Kaskel F, Update on inflammation in chronic kidney disease, Blood Purif 39 (2015) 84–92. 10.1159/000368940. [DOI] [PubMed] [Google Scholar]
- [107].Chung MK, Martin DO, Sprecher D, Wazni O, Kanderian A, Carnes CA, Bauer JA, Tchou PJ, Niebauer MJ, Natale A, Van Wagoner DR, C-reactive protein elevation in patients with atrial arrhythmias: inflammatory mechanisms and persistence of atrial fibrillation, Circulation 104 (2001) 2886–91. 10.1161/hc4901.101760. [DOI] [PubMed] [Google Scholar]
- [108].Liu C, Chu D, Kalantar-Zadeh K, George J, Young HA, Liu G, Cytokines: From Clinical Significance to Quantification, Adv Sci (Weinh) 8 (2021) e2004433. 10.1002/advs.202004433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Henein MY, Vancheri S, Longo G, Vancheri F, The Role of Inflammation in Cardiovascular Disease, Int J Mol Sci 23 (2022). 10.3390/ijms232112906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [110].Chen PS, Tan AY, Autonomic nerve activity and atrial fibrillation, Heart Rhythm 4 (2007) S61–4. 10.1016/j.hrthm.2006.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [111].Nattel S, Burstein B, Dobrev D, Atrial remodeling and atrial fibrillation: mechanisms and implications, Circ Arrhythm Electrophysiol 1 (2008) 62–73. 10.1161/CIRCEP.107.754564. [DOI] [PubMed] [Google Scholar]
- [112].Heinrich PC, Behrmann I, Muller-Newen G, Schaper F, Graeve L, Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway, Biochem J 334 (Pt 2) (1998) 297–314. 10.1042/bj3340297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Rose-John S, IL-6 trans-signaling via the soluble IL-6 receptor: importance for the pro-inflammatory activities of IL-6, Int J Biol Sci 8 (2012) 1237–47. 10.7150/ijbs.4989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Mullberg J, Schooltink H, Stoyan T, Gunther M, Graeve L, Buse G, Mackiewicz A, Heinrich PC, Rose-John S, The soluble interleukin-6 receptor is generated by shedding, Eur J Immunol 23 (1993) 473–80. 10.1002/eji.1830230226. [DOI] [PubMed] [Google Scholar]
- [115].Tsai CT, Lai LP, Kuo KT, Hwang JJ, Hsieh CS, Hsu KL, Tseng CD, Tseng YZ, Chiang FT, Lin JL, Angiotensin II activates signal transducer and activators of transcription 3 via Rac1 in atrial myocytes and fibroblasts: implication for the therapeutic effect of statin in atrial structural remodeling, Circulation 117 (2008) 344–55. 10.1161/CIRCULATIONAHA.107.695346. [DOI] [PubMed] [Google Scholar]
- [116].Sano M, Fukuda K, Kodama H, Pan J, Saito M, Matsuzaki J, Takahashi T, Makino S, Kato T, Ogawa S, Interleukin-6 family of cytokines mediate angiotensin II-induced cardiac hypertrophy in rodent cardiomyocytes, J Biol Chem 275 (2000) 29717–23. 10.1074/jbc.M003128200. [DOI] [PubMed] [Google Scholar]
- [117].Schlaich MP, Socratous F, Hennebry S, Eikelis N, Lambert EA, Straznicky N, Esler MD, Lambert GW, Sympathetic activation in chronic renal failure, J Am Soc Nephrol 20 (2009) 933–9. 10.1681/ASN.2008040402. [DOI] [PubMed] [Google Scholar]
- [118].Dziemidowicz M, Bonda TA, Litvinovich S, Taranta A, Winnicka MM, Kaminski KA, The role of interleukin-6 in intracellular signal transduction after chronic beta-adrenergic stimulation in mouse myocardium, Arch Med Sci 15 (2019) 1565–1575. 10.5114/aoms.2019.89452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Amdur RL, Feldman HI, Gupta J, Yang W, Kanetsky P, Shlipak M, Rahman M, Lash JP, Townsend RR, Ojo A, Roy-Chaudhury A, Go AS, Joffe M, He J, Balakrishnan VS, Kimmel PL, Kusek JW, Raj DS, Investigators CS, Inflammation and Progression of CKD: The CRIC Study, Clin J Am Soc Nephrol 11 (2016) 1546–1556. 10.2215/CJN.13121215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [120].Amdur RL, Mukherjee M, Go A, Barrows IR, Ramezani A, Shoji J, Reilly MP, Gnanaraj J, Deo R, Roas S, Keane M, Master S, Teal V, Soliman EZ, Yang P, Feldman H, Kusek JW, Tracy CM, Raj DS, Investigators CS, Interleukin-6 Is a Risk Factor for Atrial Fibrillation in Chronic Kidney Disease: Findings from the CRIC Study, PLoS One 11 (2016) e0148189. 10.1371/journal.pone.0148189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [121].Barreto DV, Barreto FC, Liabeuf S, Temmar M, Lemke HD, Tribouilloy C, Choukroun G, Vanholder R, Massy ZA, European G Uremic Toxin Work, Plasma interleukin-6 is independently associated with mortality in both hemodialysis and pre-dialysis patients with chronic kidney disease, Kidney Int 77 (2010) 550–6. 10.1038/ki.2009.503. [DOI] [PubMed] [Google Scholar]
- [122].Ganz T, Anemia of Inflammation, N Engl J Med 381 (2019) 1148–1157. 10.1056/NEJMra1804281. [DOI] [PubMed] [Google Scholar]
- [123].Pergola PE, Devalaraja M, Fishbane S, Chonchol M, Mathur VS, Smith MT, Lo L, Herzog K, Kakkar R, Davidson MH, Ziltivekimab for Treatment of Anemia of Inflammation in Patients on Hemodialysis: Results from a Phase 1/2 Multicenter, Randomized, Double-Blind, Placebo-Controlled Trial, J Am Soc Nephrol 32 (2021) 211–222. 10.1681/ASN.2020050595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Durlacher-Betzer K, Hassan A, Levi R, Axelrod J, Silver J, Naveh-Many T, Interleukin-6 contributes to the increase in fibroblast growth factor 23 expression in acute and chronic kidney disease, Kidney Int 94 (2018) 315–325. 10.1016/j.kint.2018.02.026. [DOI] [PubMed] [Google Scholar]
- [125].Shimada T, Kakitani M, Yamazaki Y, Hasegawa H, Takeuchi Y, Fujita T, Fukumoto S, Tomizuka K, Yamashita T, Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism, J Clin Invest 113 (2004) 561–8. 10.1172/JCI19081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [126].Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, Goetz R, Kuro-o M, Mohammadi M, Sirkis R, Naveh-Many T, Silver J, The parathyroid is a target organ for FGF23 in rats, J Clin Invest 117 (2007) 4003–8. 10.1172/JCI32409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [127].Leifheit-Nestler M, Kirchhoff F, Nespor J, Richter B, Soetje B, Klintschar M, Heineke J, Haffner D, Fibroblast growth factor 23 is induced by an activated renin-angiotensin-aldosterone system in cardiac myocytes and promotes the pro-fibrotic crosstalk between cardiac myocytes and fibroblasts, Nephrol Dial Transplant 33 (2018) 1722–1734. 10.1093/ndt/gfy006. [DOI] [PubMed] [Google Scholar]
- [128].Chen W, Yuan H, Cao W, Wang T, Chen W, Yu H, Fu Y, Jiang B, Zhou H, Guo H, Zhao X, Blocking interleukin-6 trans-signaling protects against renal fibrosis by suppressing STAT3 activation, Theranostics 9 (2019) 3980–3991. 10.7150/thno.32352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [129].Huang Z, Chen XJ, Qian C, Dong Q, Ding D, Wu QF, Li J, Wang HF, Li WH, Xie Q, Cheng X, Zhao N, Du YM, Liao YH, Signal Transducer and Activator of Transcription 3/MicroRNA-21 Feedback Loop Contributes to Atrial Fibrillation by Promoting Atrial Fibrosis in a Rat Sterile Pericarditis Model, Circ Arrhythm Electrophysiol 9 (2016) e003396. 10.1161/CIRCEP.115.003396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [130].Popa C, Netea MG, van Riel PL, van der Meer JW, Stalenhoef AF, The role of TNF-alpha in chronic inflammatory conditions, intermediary metabolism, and cardiovascular risk, J Lipid Res 48 (2007) 751–62. 10.1194/jlr.R600021-JLR200. [DOI] [PubMed] [Google Scholar]
- [131].Sonkar GK, Usha, Singh RG, Evaluation of serum tumor necrosis factor alpha and its correlation with histology in chronic kidney disease, stable renal transplant and rejection cases, Saudi J Kidney Dis Transpl 20 (2009) 1000–4. [PubMed] [Google Scholar]
- [132].Therrien FJ, Agharazii M, Lebel M, Lariviere R, Neutralization of tumor necrosis factor-alpha reduces renal fibrosis and hypertension in rats with renal failure, Am J Nephrol 36 (2012) 151–61. 10.1159/000340033. [DOI] [PubMed] [Google Scholar]
- [133].Awad AS, You H, Gao T, Cooper TK, Nedospasov SA, Vacher J, Wilkinson PF, Farrell FX, Brian Reeves W, Macrophage-derived tumor necrosis factor-alpha mediates diabetic renal injury, Kidney Int 88 (2015) 722–33. 10.1038/ki.2015.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [134].Wu N, Xu B, Xiang Y, Wu L, Zhang Y, Ma X, Tong S, Shu M, Song Z, Li Y, Zhong L, Association of inflammatory factors with occurrence and recurrence of atrial fibrillation: a meta-analysis, Int J Cardiol 169 (2013) 62–72. 10.1016/j.ijcard.2013.08.078. [DOI] [PubMed] [Google Scholar]
- [135].Qu YC, Du YM, Wu SL, Chen QX, Wu HL, Zhou SF, Activated nuclear factor-kappaB and increased tumor necrosis factor-alpha in atrial tissue of atrial fibrillation, Scand Cardiovasc J 43 (2009) 292–7. 10.1080/14017430802651803. [DOI] [PubMed] [Google Scholar]
- [136].Turner NA, Mughal RS, Warburton P, O’Regan DJ, Ball SG, Porter KE, Mechanism of TNFalpha-induced IL-1alpha, IL-1beta and IL-6 expression in human cardiac fibroblasts: effects of statins and thiazolidinediones, Cardiovasc Res 76 (2007) 81–90. 10.1016/j.cardiores.2007.06.003. [DOI] [PubMed] [Google Scholar]
- [137].Liew R, Khairunnisa K, Gu Y, Tee N, Yin NO, Naylynn TM, Moe KT, Role of tumor necrosis factor-alpha in the pathogenesis of atrial fibrosis and development of an arrhythmogenic substrate, Circ J 77 (2013) 1171–9. 10.1253/circj.cj-12-1155. [DOI] [PubMed] [Google Scholar]
- [138].Saba S, Janczewski AM, Baker LC, Shusterman V, Gursoy EC, Feldman AM, Salama G, McTiernan CF, London B, Atrial contractile dysfunction, fibrosis, and arrhythmias in a mouse model of cardiomyopathy secondary to cardiac-specific overexpression of tumor necrosis factor-alpha, Am J Physiol Heart Circ Physiol 289 (2005) H1456–67. 10.1152/ajpheart.00733.2004. [DOI] [PubMed] [Google Scholar]
- [139].Keefe JA, Hulsurkar MM, Reilly S, Wehrens XHT, Mouse models of spontaneous atrial fibrillation, Mamm Genome 34 (2023) 298–311. 10.1007/s00335-022-09964-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [140].Buckley LF, Canada JM, Carbone S, Trankle CR, Kadariya D, Billingsley H, Wohlford GF, Kirkman DL, Abbate A, Van Tassell BW, Potential role for interleukin-1 in the cardio-renal syndrome, Eur J Heart Fail 21 (2019) 385–386. 10.1002/ejhf.1403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [141].Ridker PM, MacFadyen JG, Glynn RJ, Koenig W, Libby P, Everett BM, Lefkowitz M, Thuren T, Cornel JH, Inhibition of Interleukin-1beta by Canakinumab and Cardiovascular Outcomes in Patients With Chronic Kidney Disease, J Am Coll Cardiol 71 (2018) 2405–2414. 10.1016/j.jacc.2018.03.490. [DOI] [PubMed] [Google Scholar]
- [142].Heijman J, Muna AP, Veleva T, Molina CE, Sutanto H, Tekook M, Wang Q, Abu-Taha IH, Gorka M, Kunzel S, El-Armouche A, Reichenspurner H, Kamler M, Nikolaev V, Ravens U, Li N, Nattel S, Wehrens XHT, Dobrev D, Atrial Myocyte NLRP3/CaMKII Nexus Forms a Substrate for Postoperative Atrial Fibrillation, Circ Res 127 (2020) 1036–1055. 10.1161/CIRCRESAHA.120.316710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [143].Chelu MG, Sarma S, Sood S, Wang S, van Oort RJ, Skapura DG, Li N, Santonastasi M, Muller FU, Schmitz W, Schotten U, Anderson ME, Valderrabano M, Dobrev D, Wehrens XH, Calmodulin kinase II-mediated sarcoplasmic reticulum Ca2+ leak promotes atrial fibrillation in mice, J Clin Invest 119 (2009) 1940–51. 10.1172/jci37059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [144].Tosato G, Jones KD, Interleukin-1 induces interleukin-6 production in peripheral blood monocytes, Blood 75 (1990) 1305–10. [PubMed] [Google Scholar]
- [145].Wu Q, Liu H, Liao J, Zhao N, Tse G, Han B, Chen L, Huang Z, Du Y, Colchicine prevents atrial fibrillation promotion by inhibiting IL-1beta-induced IL-6 release and atrial fibrosis in the rat sterile pericarditis model, Biomed Pharmacother 129 (2020) 110384. 10.1016/j.biopha.2020.110384. [DOI] [PubMed] [Google Scholar]
- [146].Yaribeygi H, Atkin SL, Sahebkar A, Interleukin-18 and diabetic nephropathy: A review, J Cell Physiol 234 (2019) 5674–5682. 10.1002/jcp.27427. [DOI] [PubMed] [Google Scholar]
- [147].Dai SM, Matsuno H, Nakamura H, Nishioka K, Yudoh K, Interleukin-18 enhances monocyte tumor necrosis factor alpha and interleukin-1beta production induced by direct contact with T lymphocytes: implications in rheumatoid arthritis, Arthritis Rheum 50 (2004) 432–43. 10.1002/art.20064. [DOI] [PubMed] [Google Scholar]
- [148].Miyauchi K, Takiyama Y, Honjyo J, Tateno M, Haneda M, Upregulated IL-18 expression in type 2 diabetic subjects with nephropathy: TGF-beta1 enhanced IL-18 expression in human renal proximal tubular epithelial cells, Diabetes Res Clin Pract 83 (2009) 190–9. 10.1016/j.diabres.2008.11.018. [DOI] [PubMed] [Google Scholar]
- [149].Elmarakby AA, Sullivan JC, Relationship between oxidative stress and inflammatory cytokines in diabetic nephropathy, Cardiovasc Ther 30 (2012) 49–59. 10.1111/j.1755-5922.2010.00218.x. [DOI] [PubMed] [Google Scholar]
- [150].Morel JC, Park CC, Woods JM, Koch AE, A novel role for interleukin-18 in adhesion molecule induction through NF kappa B and phosphatidylinositol (PI) 3-kinase-dependent signal transduction pathways, J Biol Chem 276 (2001) 37069–75. 10.1074/jbc.M103574200. [DOI] [PubMed] [Google Scholar]
- [151].Stuyt RJ, Netea MG, Geijtenbeek TB, Kullberg BJ, Dinarello CA, van der Meer JW, Selective regulation of intercellular adhesion molecule-1 expression by interleukin-18 and interleukin-12 on human monocytes, Immunology 110 (2003) 329–34. 10.1046/j.1365-2567.2003.01747.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [152].Maida CD, Vasto S, Di Raimondo D, Casuccio A, Vassallo V, Daidone M, Del Cuore A, Pacinella G, Cirrincione A, Simonetta I, Della Corte V, Rizzica S, Geraci G, Tuttolomondo A, Pinto A, Inflammatory activation and endothelial dysfunction markers in patients with permanent atrial fibrillation: a cross-sectional study, Aging (Albany NY) 12 (2020) 8423–8433. 10.18632/aging.103149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [153].Luan Y, Guo Y, Li S, Yu B, Zhu S, Li S, Li N, Tian Z, Peng C, Cheng J, Li Q, Cui J, Tian Y, Interleukin-18 among atrial fibrillation patients in the absence of structural heart disease, Europace 12 (2010) 1713–8. 10.1093/europace/euq321. [DOI] [PubMed] [Google Scholar]
- [154].Liu Y, Luo D, Liu E, Liu T, Xu G, Liang X, Yuan M, Zhang Y, Chen X, Chen X, Miao S, Shangguan W, Li G, MiRNA21 and IL-18 levels in left atrial blood in patients with atrial fibrillation undergoing cryoablation and their predictive value for recurrence of atrial fibrillation, J Interv Card Electrophysiol 64 (2022) 111–120. 10.1007/s10840-022-01125-z. [DOI] [PubMed] [Google Scholar]
- [155].Keefe JA, Navarro-Garcia JA, Ni L, Reilly S, Dobrev D, Wehrens XHT, In-depth characterization of a mouse model of postoperative atrial fibrillation, J Cardiovasc Aging 2 (2022). 10.20517/jca.2022.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [156].Song J, Navarro-Garcia JA, Wu J, Saljic A, Abu-Taha I, Li L, Lahiri SK, Keefe JA, Aguilar-Sanchez Y, Moore OM, Yuan Y, Wang X, Kamler M, Mitch WE, Ruiz-Hurtado G, Hu Z, Thomas SS, Dobrev D, Wehrens XH, Li N, Chronic kidney disease promotes atrial fibrillation via inflammasome pathway activation, J Clin Invest 133 (2023). 10.1172/JCI167517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [157].Cortvrindt C, Speeckaert R, Moerman A, Delanghe JR, Speeckaert MM, The role of interleukin-17A in the pathogenesis of kidney diseases, Pathology 49 (2017) 247–258. 10.1016/j.pathol.2017.01.003. [DOI] [PubMed] [Google Scholar]
- [158].Chung BH, Kim KW, Sun IO, Choi SR, Park HS, Jeon EJ, Kim BM, Choi BS, Park CW, Kim YS, Cho ML, Yang CW, Increased interleukin-17 producing effector memory T cells in the end-stage renal disease patients, Immunol Lett 141 (2012) 181–9. 10.1016/j.imlet.2011.10.002. [DOI] [PubMed] [Google Scholar]
- [159].Lang CL, Wang MH, Hung KY, Hsu SH, Chiang CK, Lu KC, Correlation of interleukin-17-producing effector memory T cells and CD4+CD25+Foxp3 regulatory T cells with the phosphate levels in chronic hemodialysis patients, ScientificWorldJournal 2014 (2014) 593170. 10.1155/2014/593170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [160].Dedong H, Feiyan Z, Jie S, Xiaowei L, Shaoyang W, Analysis of interleukin-17 and interleukin-23 for estimating disease activity and predicting the response to treatment in active lupus nephritis patients, Immunol Lett 210 (2019) 33–39. 10.1016/j.imlet.2019.04.002. [DOI] [PubMed] [Google Scholar]
- [161].Kimura A, Kishimoto T, IL-6: regulator of Treg/Th17 balance, Eur J Immunol 40 (2010) 1830–5. 10.1002/eji.201040391. [DOI] [PubMed] [Google Scholar]
- [162].Nikoo MH, Taghavian SR, Golmoghaddam H, Arandi N, Abdi Ardakani A, Doroudchi M, Increased IL-17A in atrial fibrillation correlates with neutrophil to lymphocyte ratio, Iran J Immunol 11 (2014) 246–58. [PubMed] [Google Scholar]
- [163].Yao Y, Yang M, Liu D, Zhao Q, Immune remodeling and atrial fibrillation, Front Physiol 13 (2022) 927221. 10.3389/fphys.2022.927221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [164].Yue H, Liang W, Gu J, Zhao X, Zhang T, Qin X, Zhu G, Wu Z, Comparative transcriptome analysis to elucidate the therapeutic mechanism of colchicine against atrial fibrillation, Biomed Pharmacother 119 (2019) 109422. 10.1016/j.biopha.2019.109422. [DOI] [PubMed] [Google Scholar]
- [165].von Vietinghoff S, Koltsova EK, Mestas J, Diehl CJ, Witztum JL, Ley K, Mycophenolate mofetil decreases atherosclerotic lesion size by depression of aortic T-lymphocyte and interleukin-17-mediated macrophage accumulation, J Am Coll Cardiol 57 (2011) 2194–204. 10.1016/j.jacc.2010.12.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [166].Ogura H, Murakami M, Okuyama Y, Tsuruoka M, Kitabayashi C, Kanamoto M, Nishihara M, Iwakura Y, Hirano T, Interleukin-17 promotes autoimmunity by triggering a positive-feedback loop via interleukin-6 induction, Immunity 29 (2008) 628–36. 10.1016/j.immuni.2008.07.018. [DOI] [PubMed] [Google Scholar]
- [167].Fu XX, Zhao N, Dong Q, Du LL, Chen XJ, Wu QF, Cheng X, Du YM, Liao YH, Interleukin-17A contributes to the development of post-operative atrial fibrillation by regulating inflammation and fibrosis in rats with sterile pericarditis, Int J Mol Med 36 (2015) 83–92. 10.3892/ijmm.2015.2204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [168].Saraiva M, Vieira P, O’Garra A, Biology and therapeutic potential of interleukin-10, J Exp Med 217 (2020). 10.1084/jem.20190418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [169].Naing C, Htet NH, Basavaraj AK, Nalliah S, An association between IL-10 promoter polymorphisms and diabetic nephropathy: a meta-analysis of case-control studies, J Diabetes Metab Disord 17 (2018) 333–343. 10.1007/s40200-018-0349-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Mu W, Ouyang X, Agarwal A, Zhang L, Long DA, Cruz PE, Roncal CA, Glushakova OY, Chiodo VA, Atkinson MA, Hauswirth WW, Flotte TR, Rodriguez-Iturbe B, Johnson RJ, IL-10 suppresses chemokines, inflammation, and fibrosis in a model of chronic renal disease, J Am Soc Nephrol 16 (2005) 3651–60. 10.1681/ASN.2005030297. [DOI] [PubMed] [Google Scholar]
- [171].Kondo H, Abe I, Gotoh K, Fukui A, Takanari H, Ishii Y, Ikebe Y, Kira S, Oniki T, Saito S, Aoki K, Tanino T, Mitarai K, Kawano K, Miyoshi M, Fujinami M, Yoshimura S, Ayabe R, Okada N, Nagano Y, Akioka H, Shinohara T, Akiyoshi K, Masaki T, Teshima Y, Yufu K, Nakagawa M, Takahashi N, Interleukin 10 Treatment Ameliorates High-Fat Diet-Induced Inflammatory Atrial Remodeling and Fibrillation, Circ Arrhythm Electrophysiol 11 (2018) e006040. 10.1161/CIRCEP.117.006040. [DOI] [PubMed] [Google Scholar]
- [172].Kondo H, Takahashi N, Gotoh K, Fukui A, Saito S, Aoki K, Kume O, Shinohara T, Teshima Y, Saikawa T, Splenectomy exacerbates atrial inflammatory fibrosis and vulnerability to atrial fibrillation induced by pressure overload in rats: Possible role of spleen-derived interleukin-10, Heart Rhythm 13 (2016) 241–50. 10.1016/j.hrthm.2015.07.001. [DOI] [PubMed] [Google Scholar]
- [173].Ajoolabady A, Nattel S, Lip GYH, Ren J, Inflammasome Signaling in Atrial Fibrillation: JACC State-of-the-Art Review, J Am Coll Cardiol 79 (2022) 2349–2366. 10.1016/j.jacc.2022.03.379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [174].Dobrev D, Heijman J, Hiram R, Li N, Nattel S, Inflammatory signalling in atrial cardiomyocytes: a novel unifying principle in atrial fibrillation pathophysiology, Nat Rev Cardiol 20 (2023) 145–167. 10.1038/s41569-022-00759-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [175].Yao C, Veleva T, Scott L Jr., Cao S, Li L, Chen G, Jeyabal P, Pan X, Alsina KM, Abu-Taha ID, Ghezelbash S, Reynolds CL, Shen YH, LeMaire SA, Schmitz W, Muller FU, El-Armouche A, Tony Eissa N, Beeton C, Nattel S, Wehrens XHT, Dobrev D, Li N, Enhanced Cardiomyocyte NLRP3 Inflammasome Signaling Promotes Atrial Fibrillation, Circulation 138 (2018) 2227–2242. 10.1161/CIRCULATIONAHA.118.035202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [176].Chen Z, Wu C, Liu Y, Li H, Zhu Y, Huang C, Lin H, Qiao Q, Huang M, Zhu Q, Wang L, ELABELA attenuates deoxycorticosterone acetate/salt-induced hypertension and renal injury by inhibition of NADPH oxidase/ROS/NLRP3 inflammasome pathway, Cell Death Dis 11 (2020) 698. 10.1038/s41419-020-02912-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [177].Lin Q, Li S, Jiang N, Jin H, Shao X, Zhu X, Wu J, Zhang M, Zhang Z, Shen J, Zhou W, Gu L, Lu R, Ni Z, Inhibiting NLRP3 inflammasome attenuates apoptosis in contrast-induced acute kidney injury through the upregulation of HIF1A and BNIP3-mediated mitophagy, Autophagy 17 (2021) 2975–2990. 10.1080/15548627.2020.1848971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Wen Y, Pan MM, Lv LL, Tang TT, Zhou LT, Wang B, Liu H, Wang FM, Ma KL, Tang RN, Liu BC, Artemisinin attenuates tubulointerstitial inflammation and fibrosis via the NF-kappaB/NLRP3 pathway in rats with 5/6 subtotal nephrectomy, J Cell Biochem 120 (2019) 4291–4300. 10.1002/jcb.27714. [DOI] [PubMed] [Google Scholar]
- [179].Qiu H, Ji C, Liu W, Wu Y, Lu Z, Lin Q, Xue Z, Liu X, Wu H, Jiang W, Zou C, Chronic Kidney Disease Increases Atrial Fibrillation Inducibility: Involvement of Inflammation, Atrial Fibrosis, and Connexins, Front Physiol 9 (2018) 1726. 10.3389/fphys.2018.01726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [180].Wang B, Tian Y, Yin Q, AIM2 Inflammasome Assembly and Signaling, Adv Exp Med Biol 1172 (2019) 143–155. 10.1007/978-981-13-9367-9_7. [DOI] [PubMed] [Google Scholar]
- [181].Sharma BR, Karki R, Kanneganti TD, Role of AIM2 inflammasome in inflammatory diseases, cancer and infection, Eur J Immunol 49 (2019) 1998–2011. 10.1002/eji.201848070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [182].Komada T, Chung H, Lau A, Platnich JM, Beck PL, Benediktsson H, Duff HJ, Jenne CN, Muruve DA, Macrophage Uptake of Necrotic Cell DNA Activates the AIM2 Inflammasome to Regulate a Proinflammatory Phenotype in CKD, J Am Soc Nephrol 29 (2018) 1165–1181. 10.1681/ASN.2017080863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [183].Lusebrink E, Goody PR, Lahrmann C, Flender A, Niepmann ST, Zietzer A, Schulz C, Massberg S, Jansen F, Nickenig G, Zimmer S, Krogmann AO, AIM2 Stimulation Impairs Reendothelialization and Promotes the Development of Atherosclerosis in Mice, Front Cardiovasc Med 7 (2020) 582482. 10.3389/fcvm.2020.582482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [184].Zhang W, Yan G, Xu C, Tang C, The Association of Serum AIM2 Level with the Prediction and Short-Term Prognosis of Coronary Artery Disease, J Renin Angiotensin Aldosterone Syst 2022 (2022) 6774416. 10.1155/2022/6774416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [185].Chai D, Yue Y, Xu W, Dong C, Xiong S, Mucosal co-immunization with AIM2 enhances protective SIgA response and increases prophylactic efficacy of chitosan-DNA vaccine against coxsackievirus B3-induced myocarditis, Hum Vaccin Immunother 10 (2014) 1284–94. 10.4161/hv.28333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [186].Onodi Z, Ruppert M, Kucsera D, Sayour AA, Toth VE, Koncsos G, Novak J, Brenner GB, Makkos A, Baranyai T, Giricz Z, Gorbe A, Leszek P, Gyongyosi M, Horvath IG, Schulz R, Merkely B, Ferdinandy P, Radovits T, Varga ZV, AIM2-driven inflammasome activation in heart failure, Cardiovasc Res 117 (2021) 2639–2651. 10.1093/cvr/cvab202. [DOI] [PubMed] [Google Scholar]
- [187].Song J, Wu J, Robichaux DJ, Li T, Wang S, Arredondo Sancristobal MJ, Dong B, Dobrev D, Karch J, Thomas SS, Li N, A High-Protein Diet Promotes Atrial Arrhythmogenesis via Absent-in-Melanoma 2 Inflammasome, Cells 13 (2024). 10.3390/cells13020108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [188].Li X, Ma C, Dong J, Liu X, Long D, Tian Y, Yu R, The fibrosis and atrial fibrillation: is the transforming growth factor-beta 1 a candidate etiology of atrial fibrillation, Med Hypotheses 70 (2008) 317–9. 10.1016/j.mehy.2007.04.046. [DOI] [PubMed] [Google Scholar]
- [189].Verheule S, Sato T, Everett T.t., Engle SK, Otten D, Rubart-von der Lohe M, Nakajima HO, Nakajima H, Field LJ, Olgin JE, Increased vulnerability to atrial fibrillation in transgenic mice with selective atrial fibrosis caused by overexpression of TGF-beta1, Circ Res 94 (2004) 1458–65. 10.1161/01.RES.0000129579.59664.9d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [190].Nakajima H, Nakajima HO, Salcher O, Dittie AS, Dembowsky K, Jing S, Field LJ, Atrial but not ventricular fibrosis in mice expressing a mutant transforming growth factor-beta(1) transgene in the heart, Circ Res 86 (2000) 571–9. 10.1161/01.res.86.5.571. [DOI] [PubMed] [Google Scholar]
- [191].Gu YY, Liu XS, Huang XR, Yu XQ, Lan HY, Diverse Role of TGF-beta in Kidney Disease, Front Cell Dev Biol 8 (2020) 123. 10.3389/fcell.2020.00123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [192].Mou X, Zhou DY, Zhou DY, Ma JR, Liu YH, Chen HP, Hu YB, Shou CM, Chen JW, Liu WH, Ma GL, Serum TGF-beta1 as a Biomarker for Type 2 Diabetic Nephropathy: A Meta-Analysis of Randomized Controlled Trials, PLoS One 11 (2016) e0149513. 10.1371/journal.pone.0149513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [193].Huang XR, Chung AC, Wang XJ, Lai KN, Lan HY, Mice overexpressing latent TGF-beta1 are protected against renal fibrosis in obstructive kidney disease, Am J Physiol Renal Physiol 295 (2008) F118–27. 10.1152/ajprenal.00021.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [194].Voelker J, Berg PH, Sheetz M, Duffin K, Shen T, Moser B, Greene T, Blumenthal SS, Rychlik I, Yagil Y, Zaoui P, Lewis JB, Anti-TGF-beta1 Antibody Therapy in Patients with Diabetic Nephropathy, J Am Soc Nephrol 28 (2017) 953–962. 10.1681/ASN.2015111230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [195].Trachtman H, Fervenza FC, Gipson DS, Heering P, Jayne DR, Peters H, Rota S, Remuzzi G, Rump LC, Sellin LK, Heaton JP, Streisand JB, Hard ML, Ledbetter SR, Vincenti F, A phase 1, single-dose study of fresolimumab, an anti-TGF-beta antibody, in treatment-resistant primary focal segmental glomerulosclerosis, Kidney Int 79 (2011) 1236–43. 10.1038/ki.2011.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [196].Vincenti F, Fervenza FC, Campbell KN, Diaz M, Gesualdo L, Nelson P, Praga M, Radhakrishnan J, Sellin L, Singh A, Thornley-Brown D, Veronese FV, Accomando B, Engstrand S, Ledbetter S, Lin J, Neylan J, Tumlin J, Focal G Segmental Glomerulosclerosis Study, A Phase 2, Double-Blind, Placebo-Controlled, Randomized Study of Fresolimumab in Patients With Steroid-Resistant Primary Focal Segmental Glomerulosclerosis, Kidney Int Rep 2 (2017) 800–810. 10.1016/j.ekir.2017.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [197].Hester TR Jr., Gerut ZE, Shire JR, Nguyen DB, Chen AH, Diamond J, Desmond JC, Silvati-Fidell L, Abrams SZ, Exploratory, randomized, controlled, phase 2 study to evaluate the safety and efficacy of adjuvant fibrin sealant VH S/D 4 S-Apr (ARTISS) in patients undergoing rhytidectomy, Aesthet Surg J 33 (2013) 323–33. 10.1177/1090820X13477860. [DOI] [PubMed] [Google Scholar]
- [198].You YK, Luo Q, Wu WF, Zhang JJ, Zhu HJ, Lao L, Lan HY, Chen HY, Cheng YX, Petchiether A attenuates obstructive nephropathy by suppressing TGF-beta/Smad3 and NF-kappaB signalling, J Cell Mol Med 23 (2019) 5576–5587. 10.1111/jcmm.14454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [199].Liu Y, Niu XH, Yin X, Liu YJ, Han C, Yang J, Huang X, Yu X, Gao L, Yang YZ, Xia YL, Li HH, Elevated Circulating Fibrocytes Is a Marker of Left Atrial Fibrosis and Recurrence of Persistent Atrial Fibrillation, J Am Heart Assoc 7 (2018). 10.1161/JAHA.117.008083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [200].Soppert J, Frisch J, Wirth J, Hemmers C, Boor P, Kramann R, Vondenhoff S, Moellmann J, Lehrke M, Hohl M, van der Vorst EPC, Werner C, Speer T, Maack C, Marx N, Jankowski J, Roma LP, Noels H, A systematic review and meta-analysis of murine models of uremic cardiomyopathy, Kidney Int 101 (2022) 256–273. 10.1016/j.kint.2021.10.025. [DOI] [PubMed] [Google Scholar]
- [201].Bao YW, Yuan Y, Chen JH, Lin WQ, Kidney disease models: tools to identify mechanisms and potential therapeutic targets, Zool Res 39 (2018) 72–86. 10.24272/j.issn.2095-8137.2017.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [202].Kulkarni N, Gukathasan N, Sartori S, Baber U, Chronic Kidney Disease and Atrial Fibrillation: A Contemporary Overview, J Atr Fibrillation 5 (2012) 448. 10.4022/jafib.448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [203].Lentine KL, Schnitzler MA, Abbott KC, Li L, Xiao H, Burroughs TE, Takemoto SK, Willoughby LM, Gavard JA, Brennan DC, Incidence, predictors, and associated outcomes of atrial fibrillation after kidney transplantation, Clin J Am Soc Nephrol 1 (2006) 288–96. 10.2215/CJN.00920805. [DOI] [PubMed] [Google Scholar]
- [204].Lenihan CR, Montez-Rath ME, Scandling JD, Turakhia MP, Winkelmayer WC, Outcomes after kidney transplantation of patients previously diagnosed with atrial fibrillation, Am J Transplant 13 (2013) 1566–75. 10.1111/ajt.12197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [205].Zebe H, Atrial fibrillation in dialysis patients, Nephrology Dialysis Transplantation 15 (2000) 765–768. [DOI] [PubMed] [Google Scholar]
- [206].Ting SM, Lee D, Maclean D, Sheerin NS, Paranoid psychosis and myoclonus: flecainide toxicity in renal failure, Cardiology 111 (2008) 83–6. 10.1159/000119694. [DOI] [PubMed] [Google Scholar]
- [207].Naruse Y, Tada H, Sekiguchi Y, Machino T, Ozawa M, Yamasaki H, Igarashi M, Kuroki K, Itoh Y, Murakoshi N, Yamaguchi I, Aonuma K, Concomitant chronic kidney disease increases the recurrence of atrial fibrillation after catheter ablation of atrial fibrillation: a mid-term follow-up, Heart Rhythm 8 (2011) 335–41. 10.1016/j.hrthm.2010.10.047. [DOI] [PubMed] [Google Scholar]
- [208].Sairaku A, Yoshida Y, Kamiya H, Tatematsu Y, Nanasato M, Hirayama H, Nakano Y, Kihara Y, Outcomes of ablation of paroxysmal atrial fibrillation in patients on chronic hemodialysis, J Cardiovasc Electrophysiol 23 (2012) 1289–94. 10.1111/j.1540-8167.2012.02422.x. [DOI] [PubMed] [Google Scholar]
- [209].Williams ES, Thompson VP, Chiswell KE, Alexander JH, White HD, Ohman EM, Al-Khatib SM, Rate versus rhythm control and outcomes in patients with atrial fibrillation and chronic kidney disease: data from the GUSTO-III Trial, Cardiol J 20 (2013) 439–46. 10.5603/CJ.2013.0104. [DOI] [PubMed] [Google Scholar]
- [210].Levy S, Breithardt G, Campbell R, Camm A, Daubert J-C, Allessie M, Aliot E, Capucci A, Cosio F, Crijns H, Atrial fibrillation: current knowledge and recommendations for management, European heart journal 19 (1998) 1294–1320. [DOI] [PubMed] [Google Scholar]
- [211].Reinecke H, Nabauer M, Gerth A, Limbourg T, Treszl A, Engelbertz C, Eckardt L, Kirchhof P, Wegscheider K, Ravens U, Morbidity and treatment in patients with atrial fibrillation and chronic kidney disease, Kidney international 87 (2015) 200–209. [DOI] [PubMed] [Google Scholar]
- [212].Chan KE, Lazarus JM, Thadhani R, Hakim RM, Warfarin use associates with increased risk for stroke in hemodialysis patients with atrial fibrillation, J Am Soc Nephrol 20 (2009) 2223–33. 10.1681/ASN.2009030319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [213].Ma HY, Chen S, Du Y, Estrogen and estrogen receptors in kidney diseases, Ren Fail 43 (2021) 619–642. 10.1080/0886022X.2021.1901739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [214].Rojas R, Clegg DJ, Palmer BF, Amenorrhea and Estrogen Disorders in Kidney Disease, Semin Nephrol 41 (2021) 126–132. 10.1016/j.semnephrol.2021.03.007. [DOI] [PubMed] [Google Scholar]
- [215].Wong JA, Rexrode KM, Sandhu RK, Moorthy MV, Conen D, Albert CM, Menopausal age, postmenopausal hormone therapy and incident atrial fibrillation, Heart 103 (2017) 1954–1961. 10.1136/heartjnl-2016-311002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [216].Manson JE, Hsia J, Johnson KC, Rossouw JE, Assaf AR, Lasser NL, Trevisan M, Black HR, Heckbert SR, Detrano R, Strickland OL, Wong ND, Crouse JR, Stein E, Cushman M, Women’s Health Initiative I, Estrogen plus progestin and the risk of coronary heart disease, N Engl J Med 349 (2003) 523–34. 10.1056/NEJMoa030808. [DOI] [PubMed] [Google Scholar]
- [217].Bukowska A, Spiller L, Wolke C, Lendeckel U, Weinert S, Hoffmann J, Bornfleth P, Kutschka I, Gardemann A, Isermann B, Goette A, Protective regulation of the ACE2/ACE gene expression by estrogen in human atrial tissue from elderly men, Exp Biol Med (Maywood) 242 (2017) 1412–1423. 10.1177/1535370217718808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [218].Babiker FA, De Windt LJ, van Eickels M, Grohe C, Meyer R, Doevendans PA, Estrogenic hormone action in the heart: regulatory network and function, Cardiovasc Res 53 (2002) 709–19. 10.1016/s0008-6363(01)00526-0. [DOI] [PubMed] [Google Scholar]
- [219].Wells SP, O’Shea C, Hayes S, Weeks KL, Kirchhof P, Delbridge LMD, Pavlovic D, Bell JR, Male and female atria exhibit distinct acute electrophysiological responses to sex steroids, J Mol Cell Cardiol Plus 9 (2024) 100079. 10.1016/j.jmccpl.2024.100079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [220].Ristikankare A, Kuitunen T, Kuitunen A, Uotila L, Vento A, Suojaranta-Ylinen R, Salmenpera M, Poyhia R, Lack of renoprotective effect of i.v. N-acetylcysteine in patients with chronic renal failure undergoing cardiac surgery, Br J Anaesth 97 (2006) 611–6. 10.1093/bja/ael224. [DOI] [PubMed] [Google Scholar]
- [221].Sisillo E, Ceriani R, Bortone F, Juliano G, Salvi L, Veglia F, Fiorentini C, Marenzi G, N-acetylcysteine for prevention of acute renal failure in patients with chronic renal insufficiency undergoing cardiac surgery: a prospective, randomized, clinical trial, Crit Care Med 36 (2008) 81–6. 10.1097/01.CCM.0000295305.22281.1D. [DOI] [PubMed] [Google Scholar]
- [222].Liao CY, Chung CH, Wu CC, Lin FH, Tsao CH, Wang CC, Chien WC, Protective effect of N-acetylcysteine on progression to end-stage renal disease: Necessity for prospective clinical trial, Eur J Intern Med 44 (2017) 67–73. 10.1016/j.ejim.2017.06.011. [DOI] [PubMed] [Google Scholar]
- [223].Ye M, Lin W, Zheng J, Lin S, N-acetylcysteine for chronic kidney disease: a systematic review and meta-analysis, Am J Transl Res 13 (2021) 2472–2485. [PMC free article] [PubMed] [Google Scholar]
- [224].Dember LM, Hung A, Mehrotra R, Hsu JY, Raj DS, Charytan DM, Mc Causland FR, Regunathan-Shenk R, Landis JR, Kimmel PL, Kliger AS, Himmelfarb J, Ikizler TA, Hemodialysis Novel Therapies C, A randomized controlled pilot trial of anakinra for hemodialysis inflammation, Kidney Int 102 (2022) 1178–1187. 10.1016/j.kint.2022.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [225].Rattanasompattikul M, Molnar MZ, Lee ML, Dukkipati R, Bross R, Jing J, Kim Y, Voss AC, Benner D, Feroze U, Macdougall IC, Tayek JA, Norris KC, Kopple JD, Unruh M, Kovesdy CP, Kalantar-Zadeh K, Anti-Inflammatory and Anti-Oxidative Nutrition in Hypoalbuminemic Dialysis Patients (AIONID) study: results of the pilot-feasibility, double-blind, randomized, placebo-controlled trial, J Cachexia Sarcopenia Muscle 4 (2013) 247–57. 10.1007/s13539-013-0115-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [226].Kiuchi MG, Chen S, GR ES, Rodrigues Paz LM, Kiuchi T, de Paula Filho AG, Lima Souto GL, The addition of renal sympathetic denervation to pulmonary vein isolation reduces recurrence of paroxysmal atrial fibrillation in chronic kidney disease patients, J Interv Card Electrophysiol 48 (2017) 215–222. 10.1007/s10840-016-0186-6. [DOI] [PubMed] [Google Scholar]
- [227].Steinberg JS, Shabanov V, Ponomarev D, Losik D, Ivanickiy E, Kropotkin E, Polyakov K, Ptaszynski P, Keweloh B, Yao CJ, Pokushalov EA, Romanov AB, Effect of Renal Denervation and Catheter Ablation vs Catheter Ablation Alone on Atrial Fibrillation Recurrence Among Patients With Paroxysmal Atrial Fibrillation and Hypertension: The ERADICATE-AF Randomized Clinical Trial, JAMA 323 (2020) 248–255. 10.1001/jama.2019.21187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [228].Hohl M, Selejan SR, Wintrich J, Lehnert U, Speer T, Schneider C, Mauz M, Markwirth P, Wong DWL, Boor P, Kazakov A, Mollenhauer M, Linz B, Klinkhammer BM, Hubner U, Ukena C, Moellmann J, Lehrke M, Wagenpfeil S, Werner C, Linz D, Mahfoud F, Bohm M, Renal Denervation Prevents Atrial Arrhythmogenic Substrate Development in CKD, Circ Res 130 (2022) 814–828. 10.1161/CIRCRESAHA.121.320104. [DOI] [PubMed] [Google Scholar]
- [229].Liang Z, Shi XM, Liu LF, Chen XP, Shan ZL, Lin K, Li J, Chen FK, Li YG, Guo HY, Wang YT, Renal denervation suppresses atrial fibrillation in a model of renal impairment, PLoS One 10 (2015) e0124123. 10.1371/journal.pone.0124123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [230].Konigsbrugge O, Ay C, Atrial fibrillation in patients with end-stage renal disease on hemodialysis: Magnitude of the problem and new approach to oral anticoagulation, Res Pract Thromb Haemost 3 (2019) 578–588. 10.1002/rth2.12250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [231].King BMN, Mintz S, Lin X, Morley GE, Schlamp F, Khodadadi-Jamayran A, Fishman GI, Chronic Kidney Disease Induces Proarrhythmic Remodeling, Circ Arrhythm Electrophysiol 16 (2023) e011466. 10.1161/CIRCEP.122.011466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [232].van Ham WB, Cornelissen CM, Polyakova E, van der Voorn SM, Ligtermoet ML, Monshouwer-Kloots J, Vos MA, Bossu A, van Rooij E, van der Heyden MAG, van Veen TAB, Pro-Arrhythmic Potential of Accumulated Uremic Toxins Is Mediated via Vulnerability of Action Potential Repolarization, Int J Mol Sci 24 (2023). 10.3390/ijms24065373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [233].Uchida T, Furuno Y, Tanimoto A, Toyohira Y, Arakaki K, Kina-Tanada M, Kubota H, Sakanashi M, Matsuzaki T, Noguchi K, Nakasone J, Igarashi T, Ueno S, Matsushita M, Ishiuchi S, Masuzaki H, Ohya Y, Yanagihara N, Shimokawa H, Otsuji Y, Tamura M, Tsutsui M, Development of an experimentally useful model of acute myocardial infarction: 2/3 nephrectomized triple nitric oxide synthases-deficient mouse, J Mol Cell Cardiol 77 (2014) 29–41. 10.1016/j.yjmcc.2014.09.021. [DOI] [PubMed] [Google Scholar]
- [234].Gaikwad AB, Sayyed SG, Lichtnekert J, Tikoo K, Anders HJ, Renal failure increases cardiac histone h3 acetylation, dimethylation, and phosphorylation and the induction of cardiomyopathy-related genes in type 2 diabetes, Am J Pathol 176 (2010) 1079–83. 10.2353/ajpath.2010.090528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [235].Junho CVC, Gonzalez-Lafuente L, Navarro-Garcia JA, Rodriguez-Sanchez E, Carneiro-Ramos MS, Ruiz-Hurtado G, Unilateral Acute Renal Ischemia-Reperfusion Injury Induces Cardiac Dysfunction through Intracellular Calcium Mishandling, Int J Mol Sci 23 (2022). 10.3390/ijms23042266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [236].Pang P, Abbott M, Abdi M, Fucci QA, Chauhan N, Mistri M, Proctor B, Chin M, Wang B, Yin W, Lu TS, Halim A, Lim K, Handy DE, Loscalzo J, Siedlecki AM, Pre-clinical model of severe glutathione peroxidase-3 deficiency and chronic kidney disease results in coronary artery thrombosis and depressed left ventricular function, Nephrol Dial Transplant 33 (2018) 923–934. 10.1093/ndt/gfx304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [237].Santana AC, Degaspari S, Catanozi S, Delle H, de Sa Lima L, Silva C, Blanco P, Solez K, Scavone C, Noronha IL, Thalidomide suppresses inflammation in adenine-induced CKD with uraemia in mice, Nephrol Dial Transplant 28 (2013) 1140–9. 10.1093/ndt/gfs569. [DOI] [PubMed] [Google Scholar]
- [238].Zhang W, Miikeda A, Zuckerman J, Jia X, Charugundla S, Zhou Z, Kaczor-Urbanowicz KE, Magyar C, Guo F, Wang Z, Pellegrini M, Hazen SL, Nicholas SB, Lusis AJ, Shih DM, Inhibition of microbiota-dependent TMAO production attenuates chronic kidney disease in mice, Sci Rep 11 (2021) 518. 10.1038/s41598-020-80063-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [239].Knauf F, Asplin JR, Granja I, Schmidt IM, Moeckel GW, David RJ, Flavell RA, Aronson PS, NALP3-mediated inflammation is a principal cause of progressive renal failure in oxalate nephropathy, Kidney Int 84 (2013) 895–901. 10.1038/ki.2013.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [240].Mulay SR, Eberhard JN, Pfann V, Marschner JA, Darisipudi MN, Daniel C, Romoli S, Desai J, Grigorescu M, Kumar SV, Rathkolb B, Wolf E, Hrabe de Angelis M, Bauerle T, Dietel B, Wagner CA, Amann K, Eckardt KU, Aronson PS, Anders HJ, Knauf F, Oxalate-induced chronic kidney disease with its uremic and cardiovascular complications in C57BL/6 mice, Am J Physiol Renal Physiol 310 (2016) F785–F795. 10.1152/ajprenal.00488.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [241].Nozu K, Nakanishi K, Abe Y, Udagawa T, Okada S, Okamoto T, Kaito H, Kanemoto K, Kobayashi A, Tanaka E, Tanaka K, Hama T, Fujimaru R, Miwa S, Yamamura T, Yamamura N, Horinouchi T, Minamikawa S, Nagata M, Iijima K, A review of clinical characteristics and genetic backgrounds in Alport syndrome, Clin Exp Nephrol 23 (2019) 158–168. 10.1007/s10157-018-1629-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [242].Salah SM, Meisenheimer JD, Rao R, Peda JD, Wallace DP, Foster D, Li X, Li X, Zhou X, Vallejo JA, Wacker MJ, Fields TA, Swenson-Fields KI, MCP-1 promotes detrimental cardiac physiology, pulmonary edema, and death in the cpk model of polycystic kidney disease, Am J Physiol Renal Physiol 317 (2019) F343–F360. 10.1152/ajprenal.00240.2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
